Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 34(8): 9869-9883, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32533745

RESUMO

Proteolytic cleavage of the cell adhesion molecule L1 (L1) in brain tissue and in cultured cerebellar neurons results in the generation and nuclear import of a 30 kDa fragment comprising most of L1's C-terminal, intracellular domain. In search of molecules that interact with this domain, we performed affinity chromatography with the recombinant intracellular L1 domain and a nuclear extract from mouse brains, and identified potential nuclear L1 binding partners involved in transcriptional regulation, RNA processing and transport, DNA repair, chromatin remodeling, and nucleocytoplasmic transport. By co-immunoprecipitation and enzyme-linked immunosorbent assay using recombinant proteins, we verified the direct interaction between L1 and the nuclear binding partners non-POU domain containing octamer-binding protein and splicing factor proline/glutamine-rich. The proximity ligation assay confirmed this close interaction in cultures of cerebellar granule cells. Our findings suggest that L1 fragments regulate multiple nuclear functions in the nervous system. We discuss possible physiological and pathological roles of these interactions in regulation of chromatin structure, gene expression, RNA processing, and DNA repair.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Domínios Proteicos
2.
Pediatr Res ; 89(6): 1389-1395, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32937649

RESUMO

BACKGROUND: The mechanism of bilirubin neurotoxicity is poorly understood. We hypothesize that bilirubin inhibits the function of lipid rafts (LR), microdomains of the plasma membrane critical for signal transduction. To test this hypothesis, we measured the effect of free bilirubin (Bf) between 7.6 and 122.5 nM on LR-dependent functions of L1 cell adhesion molecule (L1). METHODS: Cerebellar granule neurons (CGN) were plated on poly-L-lysine overnight, and neurite length was determined after 1 h treatment with L1 alone or L1 and bilirubin. L1 activation of ERK1/2 was measured in CGN in the presence or absence of bilirubin. The effect of bilirubin on L1 distribution in LR was quantitated, and the localization of bilirubin to LR was determined. RESULTS: The addition of bilirubin to CGN treated with L1 significantly decreased neurite length compared to L1 alone. L1 activation of ERK1/2 was inhibited by bilirubin. Bilirubin redistributed L1 into LR. Bilirubin was associated only with LR-containing fractions of a sucrose density gradient. CONCLUSION: Bf significantly inhibits LR-dependent functions of L1 and are found only associated with LR, suggesting one mechanism by which bilirubin may exert neurotoxicity is through the dysfunction of protein-LR interactions. IMPACT: This article establishes lipid rafts as a target for the neurotoxic effects of bilirubin. This article provides clear evidence toward establishing one mechanism of bilirubin neurotoxicity, where little is understood. This article paves the way for future investigation into lipid raft dependent functions, and its role in neurodevelopmental outcome.


Assuntos
Bilirrubina/farmacologia , Cerebelo/metabolismo , Grânulos Citoplasmáticos/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Animais , Ratos , Ratos Sprague-Dawley
3.
J Neurosci ; 39(34): 6656-6667, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31300520

RESUMO

The growth of axons corresponding to different neuronal subtypes is governed by unique expression profiles of molecules on the growth cone. These molecules respond to extracellular cues either locally though cell adhesion interactions or over long distances through diffusible gradients. Here, we report that that the cell adhesion molecule ALCAM (CD166) can act as an extracellular substrate to selectively promote the growth of murine midbrain dopamine (mDA) neuron axons through a trans-heterophilic interaction with mDA-bound adhesion molecules. In mixed-sex primary midbrain cultures, the growth-promoting effect of ALCAM was abolished by neutralizing antibodies for components of the Semaphorin receptor complex Nrp1, Chl1, or L1cam. The ALCAM substrate was also found to modulate the response of mDA neurites to soluble semaphorins in a context-specific manner by abolishing the growth-promoting effect of Sema3A but inducing a branching response in the presence of Sema3C. These findings identify a previously unrecognized guidance mechanism whereby cell adhesion molecules act in trans to modulate the response of axonal growth cones to soluble gradients to selectively orchestrate the growth and guidance of mDA neurons.SIGNIFICANCE STATEMENT The mechanisms governing the axonal connectivity of midbrain dopamine (mDA) neurons during neural development have remained rather poorly understood relative to other model systems for axonal growth and guidance. Here, we report a series of novel interactions between proteins previously not identified in the context of mDA neuronal growth. Significantly, the results suggest a previously unrecognized mechanism involving the convergence in signaling between local, adhesion and long-distance, soluble cues. A better understanding of the molecules and mechanisms involved in establishment of the mDA system is important as a part of ongoing efforts to understand the consequence of conditions that may result from aberrant connectivity and also for cell replacement strategies for Parkinson's disease.


Assuntos
Molécula de Adesão de Leucócito Ativado/fisiologia , Axônios/fisiologia , Moléculas de Adesão Celular/fisiologia , Neurônios Dopaminérgicos/fisiologia , Mesencéfalo/citologia , Mesencéfalo/crescimento & desenvolvimento , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Semaforinas/fisiologia , Animais , Anticorpos Bloqueadores/farmacologia , Feminino , Cones de Crescimento , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais/fisiologia
4.
Int J Mol Sci ; 21(21)2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-33167483

RESUMO

Metastasis is the main cause of death among colorectal cancer (CRC) patients. E-selectin and its carbohydrate ligands, including sialyl Lewis X (sLeX) antigen, are key players in the binding of circulating tumor cells to the endothelium, which is one of the major events leading to organ invasion. Nevertheless, the identity of the glycoprotein scaffolds presenting these glycans in CRC remains unclear. In this study, we firstly have characterized the glycoengineered cell line SW620 transfected with the fucosyltransferase 6 (FUT6) coding for the α1,3-fucosyltransferase 6 (FUT6), which is the main enzyme responsible for the synthesis of sLeX in CRC. The SW620FUT6 cell line expressed high levels of sLeX antigen and E-selectin ligands. Moreover, it displayed increased migration ability. E-selectin ligand glycoproteins were isolated from the SW620FUT6 cell line, identified by mass spectrometry, and validated by flow cytometry and Western blot (WB). The most prominent E-selectin ligand we identified was the neural cell adhesion molecule L1 (L1CAM). Previous studies have shown association of L1CAM with metastasis in cancer, thus the novel role as E-selectin counter-receptor contributes to understand the molecular mechanism involving L1CAM in metastasis formation.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Selectina E/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Adesão Celular/genética , Neoplasias do Colo/genética , Humanos , Imunoprecipitação , Ligantes , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Ligação Proteica/genética , Transfecção , Células Tumorais Cultivadas
5.
J Neurogenet ; 32(1): 15-26, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29191114

RESUMO

To study the central pattern generators functioning, previously we identified genes, whose neurospecific knockdowns led to deviations in the courtship song of Drosophila melanogaster males. Reduced expression of the gene CG15630 caused a decrease in the interpulse interval. To investigate the role of CG15630, which we have called here fipi (factor of interpulse interval), in the courtship song production, at first, we have characterized fipi transcripts and protein (FIPI) in the mutant flies carrying P insertion and deletions in this gene and in flies with its RNAi knockdown. FIPI is homologous to the mammalian NCAM2 protein, an important factor of neuronal development in the olfactory system. In this study, we have revealed that local fipi knockdown in the antennal olfactory sensory neurons (OR67d and IR84a), which are responsible for reception of chemosignals modulating courtship behavior, alters the interpulse interval in the opposite directions. Thus, a proper fipi expression seems to be necessary for perception of sexual chemosignals, and the effect of fipi knockdown on IPI value depends on the type of chemoreceptor neurons affected.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Geradores de Padrão Central/fisiologia , Masculino
6.
Int J Med Sci ; 14(12): 1276-1283, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29104485

RESUMO

Background: Cell recognition molecule L1 (L1) plays an important role in cancer cell differentiation, proliferation, migration and survival, but its mechanism remains unclear. Methodology/Principal: Our previous study has demonstrated that L1 enhanced cell survival and migration in neural cells by regulating cell surface glycosylation. In the present study, we show that L1 affected cell migration and survival in CHO (Chinese hamster ovary) cell line by modulation of sialylation and fucosylation at the cell surface via the PI3K (phosphoinositide 3-kinase) and Erk (extracellularsignal-regulated kinase) signaling pathways. Flow cytometry analysis indicated that L1 modulated cell surface sialylation and fucosylation in CHO cells. Activated L1 upregulated the protein expressions of ST6Gal1 (ß-galactoside α-2,6-sialyltransferase 1) and FUT9 (Fucosyltransferase 9) in CHO cells. Furthermore, activated L1 promoted CHO cells migration and survival as shown by transwell assay and MTT assay. Inhibitors of sialylation and fucosylation blocked L1-induced cell migration and survival, while decreasing FUT9 and ST6Gal1 expressions via the PI3K-dependent and Erk-dependent signaling pathways. Conclusion : L1 modulated cell migration and survival by regulation of cell surface sialylation and fucosylation via the PI3K-dependent and Erk-dependent signaling pathways.


Assuntos
Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Animais , Células CHO , Membrana Celular/metabolismo , Cricetulus , Fucosiltransferases/metabolismo , Glicosilação , Fosfatidilinositol 3-Quinases/metabolismo , Sialiltransferases/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(47): 16943-8, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385598

RESUMO

NGF binding to its protein kinase receptor TrkA is known to induce neurite outgrowth and neural cell differentiation. The plasma membrane expansion, necessary for the process, was shown to be contributed by the VAMP7-dependent exocytosis of endocytic vesicles. Working with wild-type PC12 (wtPC12), a cell model widely used to investigate NGF-induced neurite outgrowth, we found that a few hours of treatment with the neurotrophin (and to a lower extent with basic FGF and EGF) induces the appearance of enlargeosome vesicles competent for VAMP4-dependent exocytosis abundant in high REST-PC12 clones. Both the neurite length assay and the immunocytochemistry of enlargeosomes exocytosis revealed that activation of TrkA is induced not only by NGF, but also by the L1 adhesion protein, L1CAM, whose soluble construct binds the receptor with submicromolar affinity. In the intact wtPC12, the L1CAM construct induced autophosphorylation and internalization of TrkA followed by the activation of the PI3K, MEK, and PKCγ signaling cascades, analogous to the responses induced by NGF. Down-regulation of either VAMP7 or VAMP4 revealed the coparticipation of the two corresponding vesicles to the outgrowth responses induced by NGF and L1CAM. Finally, mixing experiments of wtPC12 cells rich in TrkA with high REST PC12 cells transfected with L1CAM documented the transactivation of the receptor by the adhesion protein surface-exposed in adjacent cells. In view of the known inhomogeneous surface distribution of both L1CAM and TrkA in various neural cells including neurons, their transcellular binding could be restricted to discrete sites, governing local signaling events distinct from those induced by soluble messengers.


Assuntos
Exocitose , Fatores de Crescimento Neural/fisiologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neuritos , Receptor trkA/agonistas , Animais , Células PC12 , Ratos
8.
Bull Exp Biol Med ; 160(6): 807-10, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27165065

RESUMO

Molecule L1CAM is specific for nerve cells and tumors of various localizations. The expression of L1CAM is significantly higher in melanoma in comparison with benign nevi and correlates with the progress of melanoma and transition from radial to vertical growth. Monoclonal antibodies to L1CAM effectively and specifically attenuate melanoma growth, though stimulates the epithelial-mesenchymal transition. shRNA-mediated knock-down of L1CAM showed the involvement of L1CAM in regulation of activity of the canonical Wnt pathway and expression of genes of class I melanoma-associated antigens (MAGE).


Assuntos
Antígenos de Neoplasias/genética , Proteínas de Neoplasias/genética , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Via de Sinalização Wnt , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo
9.
Cereb Cortex ; 24(11): 3014-24, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23780867

RESUMO

Excitatory neurons undergo dendritic spine remodeling in response to different stimuli. However, there is scarce information about this type of plasticity in interneurons. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) is a good candidate to mediate this plasticity as it participates in neuronal remodeling and is expressed by some mature cortical interneurons, which have reduced dendritic arborization, spine density, and synaptic input. To study the connectivity of the dendritic spines of interneurons and the influence of PSA-NCAM on their dynamics, we have analyzed these structures in a subpopulation of fluorescent spiny interneurons in the hippocampus of glutamic acid decarboxylase-enhanced green fluorescent protein transgenic mice. Our results show that these spines receive excitatory synapses. The depletion of PSA in vivo using the enzyme Endo-Neuraminidase-N (Endo-N) increases spine density when analyzed 2 days after, but decreases it 7 days after. The dendritic spine turnover was also analyzed in real time using organotypic hippocampal cultures: 24 h after the addition of EndoN, we observed an increase in the apparition rate of spines. These results indicate that dendritic spines are important structures in the control of the synaptic input of hippocampal interneurons and suggest that PSA-NCAM is relevant in the regulation of their morphology and connectivity.


Assuntos
Espinhas Dendríticas/metabolismo , Regulação da Expressão Gênica/fisiologia , Interneurônios/ultraestrutura , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Ácidos Siálicos/metabolismo , Ácidos Siálicos/fisiologia , Animais , Animais Recém-Nascidos , Calbindina 2/metabolismo , Colecistocinina/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Molécula L1 de Adesão de Célula Nervosa/efeitos dos fármacos , Neuraminidase/farmacologia , Técnicas de Cultura de Órgãos , Somatostatina/metabolismo , Fatores de Tempo , Peptídeo Intestinal Vasoativo/metabolismo
10.
Pathol Int ; 65(2): 58-66, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25641508

RESUMO

L1cam (L1), one of the cell adhesion molecules belonging to the immunoglobulin superfamily, plays critical roles in neuronal migration, axon growth, guidance, fasciculation, and synaptic plasticity in the central as well as the peripheral nervous system. A number of X-linked forms of mental retardation have been associated with mutations in the L1 gene, including X-linked hydrocephalus in humans. Although model mice with different sites of L1 mutation have been studied, the pathogenetic mechanisms of hydrocephalus and mental retardation still remain unsolved. We herein present an overview of the function of L1 in the central nervous system and describe a human case of L1 mutation and knock-in mice that showed deleted sixth immunoglobulin of L1. Finally, we present experimental evidence showing that L1 is involved in murine neocortical histogenesis and propose a hypothetical mechanism of L1-linked hydrocephalus, with reference to corticogenesis.


Assuntos
Encéfalo/embriologia , Hidrocefalia/fisiopatologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurogênese/fisiologia , Animais , Encéfalo/fisiologia , Modelos Animais de Doenças , Feto , Humanos , Masculino , Camundongos , Natimorto
11.
Biochem J ; 460(3): 437-46, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24673421

RESUMO

Lack of permissive mechanisms and abundance of inhibitory molecules in the lesioned central nervous system of adult mammals contribute to the failure of functional recovery, which leads to severe disabilities in motor functions or pain. Previous studies have indicated that the neural cell adhesion molecule L1 constitutes a viable target to promote regeneration. In the present study, we describe the cloning, functional expression in Escherichia coli cells and purification of a recombinant αL1 Fab fragment that binds to L1 with comparable activity as the function-triggering monoclonal antibody 557.B6 and induces neurite outgrowth and neuronal survival in cultured neurons, despite its monovalent function. Infusion of αL1 Fab into the lesioned spinal cord of mice enhanced functional recovery after thoracic spinal cord compression injury. αL1 Fab treatment resulted in reduced scar volume, enhanced number of tyrosine hydroxylase-positive axons and increased linear density of VGLUT1 (vesicular glutamate transporter 1) on motoneurons. Furthermore, the number and soma size of ChAT (choline acetyltransferase)-positive motoneurons and the linear density of ChAT-positive boutons on motoneurons as well as parvalbumin-positive interneurons in the lumbar spinal cord were elevated. Stimulation of endogenous L1 by application of the αL1 Fab opens new avenues for recombinant antibody technology, offering prospects for therapeutic applications after traumatic nervous system lesions.


Assuntos
Fragmentos Fab das Imunoglobulinas/uso terapêutico , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Colina O-Acetiltransferase/biossíntese , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/metabolismo , Regeneração Nervosa , Neuritos/fisiologia , Ratos , Proteínas Recombinantes/uso terapêutico , Traumatismos da Medula Espinal/fisiopatologia , Proteína Vesicular 1 de Transporte de Glutamato/biossíntese
12.
J Neurochem ; 128(5): 713-24, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24118054

RESUMO

The cell adhesion molecule L1 regulates cellular responses in the developing and adult nervous system. Here, we show that stimulation of cultured mouse cerebellar neurons by a function-triggering L1 antibody leads to cathepsin E-mediated generation of a sumoylated 30 kDa L1 fragment (L1-30) and to import of L1-30 into the nucleus. Mutation of the sumoylation site at K1172 or the cathepsin E cleavage site at E1167 abolishes generation of L1-30, while mutation of the nuclear localization signal at K1147 prevents nuclear import of L1-30. Moreover, the aspartyl protease inhibitor pepstatin impairs the generation of L1-30 and inhibits L1-induced migration of cerebellar neurons and Schwann cells as well as L1-dependent in vitro myelination on axons of dorsal root ganglion neurons by Schwann cells. L1-stimulated migration of HEK293 cells expressing L1 with mutated cathepsin E cleavage site is diminished in comparison to migration of cells expressing non-mutated L1. In addition, L1-stimulated migration of HEK293 cells expressing non-mutated L1 is also abolished upon knock-down of cathepsin E expression and enhanced by over-expression of cathepsin E. The findings of the present study indicate that generation and nuclear import of L1-30 regulate neuronal and Schwann cell migration as well as myelination. Cell adhesion molecule L1 regulates cellular responses in the developing and adult nervous system. L1 stimulation triggers sumoylation and cleavage of L1, thus generating the L1-70 fragment (1) which is cleaved by cathepsin E (2) yielding the L1-30 fragment that is imported to the nucleus (3), may bind to DNA and/or nuclear proteins (4), to regulate diverse cellular functions.


Assuntos
Catepsina E/metabolismo , Movimento Celular/fisiologia , Bainha de Mielina/fisiologia , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/fisiologia , Células de Schwann/fisiologia , Sumoilação/fisiologia , Animais , Axônios/efeitos dos fármacos , Axônios/fisiologia , Catepsina E/genética , Cerebelo/citologia , Técnicas de Cocultura , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Mutação/fisiologia , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neuritos/efeitos dos fármacos , Pepstatinas/farmacologia , Fragmentos de Peptídeos/metabolismo , Inibidores de Proteases/farmacologia , RNA Interferente Pequeno/genética , Sumoilação/genética
13.
Pediatr Res ; 75(1-1): 8-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24126818

RESUMO

BACKGROUND: Chlorhexidine is a skin disinfectant that reduces skin and mucous membrane bacterial colonization and inhibits organism growth. Despite numerous studies assessing chlorhexidine safety in term infants, residual concerns have limited its use in hospitalized neonates, especially low-birth-weight preterm infants. The aim of this study was to assess the potential neurotoxicity of chlorhexidine on the developing central nervous system using a well-established in vitro model of neurite outgrowth that includes laminin and L1 cell adhesion molecule (L1) as neurite outgrowth-promoting substrates. METHODS: Cerebellar granule neurons are plated on poly L-lysine, L1, or laminin. Chlorhexidine, hexachlorophene, or their excipients are added to the media. Neurons are grown for 24 h, fixed, and neurite length is measured. RESULTS: Chlorhexidine significantly reduced the length of neurites grown on L1 but not on laminin. Chlorhexidine concentrations as low as 125 ng/ml statistically significantly reduced neurite length on L1. Hexachlorophene did not affect neurite length. CONCLUSION: Chlorhexidine at concentrations detected in the blood following topical applications in preterm infants specifically inhibited L1-mediated neurite outgrowth of cerebellar granule neurons. It is now vital to determine whether the blood-brain barrier is permeable to chlorhexidine in preterm infants.


Assuntos
Clorexidina/farmacologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neuritos/efeitos dos fármacos , Humanos , Técnicas In Vitro , Recém-Nascido
14.
Alcohol Clin Exp Res ; 38(11): 2722-30, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25421509

RESUMO

BACKGROUND: Fetal alcohol spectrum disorder, the leading known cause of mental retardation, is caused by alcohol exposure during pregnancy. One mechanism of ethanol (EtOH) teratogenicity is the disruption of the functions of L1 cell adhesion molecule (L1). These functions include enhancement of neurite outgrowth, trafficking through lipid rafts, and signal transduction. Recent data have shown that choline supplementation of rat pups reduces the effects of EtOH on neurobehavior. We sought to determine whether choline could prevent the effect of EtOH on L1 function using a simple experimental system. METHODS: Cerebellar granule neurons (CGN) from postnatal day 6 rat pups were cultured with and without supplemental choline, and the effects on L1 signaling, lipid raft distribution, and neurite outgrowth were measured in the presence or absence of EtOH. RESULTS: Choline significantly reduced the effect of EtOH on L1 signaling, the distribution of L1 in lipid rafts and L1-mediated neurite outgrowth. However, choline supplemented EtOH-exposed cultures remained significantly different than controls. CONCLUSIONS: Choline pretreatment of CGN significantly reduces the disruption of L1 function by EtOH, but does not completely return L1 function to baseline. This experimental system will enable discovery of the mechanism of the neuroprotective effect of choline.


Assuntos
Colina/farmacologia , Etanol/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Relação Dose-Resposta a Droga , Etanol/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
15.
J Neurosci ; 32(11): 3917-30, 2012 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22423112

RESUMO

We have identified the adenine nucleotide translocator (ANT) isoforms ANT1 and ANT2 that are present in the plasma membrane of mouse cerebellar neurons as novel binding partners of the cell adhesion molecule L1. The direct interaction between ANT and L1 is mediated by sites within the fibronectin type III domains of L1 and the first and third extracellular loops of the ANT proteins. We also show that L1 interacts with the ANT binding partner matrix metalloprotease 14 (MMP14) and that the ANT proteins bind directly to the L1 interaction partner glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Moreover, we provide evidence that the functional interplay between L1, ANT proteins, MMP14, and GAPDH at the plasma membrane mediates L1-induced neurite outgrowth of cerebellar neurons. Disruption of this interplay by ANT inhibitors, ANT-derived synthetic peptides, and/or function-blocking MMP14 and ANT antibodies leads to alterations in L1-dependent neurite outgrowth. Stimulation of L1-mediated signaling in cerebellar neurons triggers transient ATP secretion via ANT proteins and leads to transient src family-dependent tyrosine phosphorylation of L1, ANT1, ANT2, and MMP14. Thus, our results indicate that plasma membrane-localized ANT1 and ANT2 regulate L1-mediated neurite outgrowth in conjunction with MMP14.


Assuntos
Translocador 1 do Nucleotídeo Adenina/metabolismo , Translocador 2 do Nucleotídeo Adenina/metabolismo , Cerebelo/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neuritos/fisiologia , Animais , Células Cultivadas , Cerebelo/citologia , Feminino , Masculino , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Ligação Proteica/fisiologia
16.
J Neurochem ; 124(3): 397-409, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22973895

RESUMO

The L1 syndrome, a genetic disease that affects 1/30 000 newborn males, is sustained by numerous missense mutations of L1 cell adhesion molecule (L1CAM), an adhesion surface protein active also in transmembrane signaling, essential for the development and function of neurons. To investigate the cell biology of L1CAM, we employed a high RE1-silencing transcription (factor) clone of the pheochromocytoma PC12 line, defective in L1CAM expression and neurite outgrowth. The clone was transfected with wild-type L1CAM and four missense, disease-inducing point mutants encoding proteins distributed to the cell surface. The mutant-expressing cells, defective in adhesion to extracellular matrix proteins and in migration, exhibited unchanged proliferation. The nerve growth factor (NGF)-induced neurite outgrowth was re-established in defective clone cells transfected with the wild-type and the H210Q and I219T L1CAMs mutants, but not in the others. The stimulated outgrowth was confirmed in a second defective PC12 clone over-expressing the NGF receptor TrkA, treated with NGF and/or a recombinant L1CAM chimera. These results revealed a new function of L1CAM, a positive, robust and dose-dependent modulation of the TrkA receptor activated spontaneously or by NGF. The variable effects observed with the different L1CAM mutants suggest that this function contributes to the marked heterogeneity of symptoms and severity observed in the patients affected by the L1 syndrome.


Assuntos
Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurônios/patologia , Neurônios/fisiologia , Animais , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Fator de Crescimento Neural/fisiologia , Neurônios/metabolismo , Células PC12 , Ratos , Receptor trkA/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/genética
17.
Biochem Biophys Res Commun ; 440(3): 405-12, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24070611

RESUMO

Cell recognition molecule L1 (CD171) plays an important role in neuronal survival, migration, differentiation, neurite outgrowth, myelination, synaptic plasticity and regeneration after injury. Our previous study has demonstrated that overexpressing L1 enhances cell survival and proliferation of mouse embryonic stem cells (ESCs) through promoting the expression of FUT9 and ST3Gal4, which upregulates cell surface sialylation and fucosylation. In the present study, we examined whether sialylation and fucosylation are involved in ESC differentiation through L1 signaling. RNA interference analysis showed that L1 enhanced differentiation of ESCs into neurons through the upregulation of FUT9 and ST3Gal4. Furthermore, blocking the phospholipase Cγ (PLCγ) signaling pathway with either a specific PLCγ inhibitor or knockdown PLCγ reduced the expression levels of both FUT9 and ST3Gal4 mRNAs and inhibited L1-mediated neuronal differentiation. These results demonstrate that L1 promotes neuronal differentiation from ESCs through the L1-mediated enhancement of FUT9 and ST3Gal4 expression.


Assuntos
Membrana Celular/metabolismo , Células-Tronco Embrionárias/citologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurogênese/fisiologia , Animais , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Glicosilação , Humanos , Camundongos , Molécula L1 de Adesão de Célula Nervosa/genética , Neurogênese/genética , Fosfolipase C gama/antagonistas & inibidores , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Interferência de RNA , Sialiltransferases/genética , Sialiltransferases/metabolismo , beta-Galactosídeo alfa-2,3-Sialiltransferase
18.
Cell Tissue Res ; 352(2): 387-99, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23292132

RESUMO

Cellular mechanisms induced by melatonin to synchronise seasonal reproduction in several species, including sheep, remain unclear. We sought to evaluate the scale and physiological significance of neural plasticity in order to explain the delay between the change of duration of melatonin secretion and the change of reproductive status following a transition from long days (LD, 16 h light/24 h) to short days (SD, 8 h light/24 h) and from SD to LD. Using Western blots in ovariectomised oestradiol-replaced ewes, we evaluated the content of the polysialylated form of neural cell adhesion molecule (PSA-NCAM), a plasticity marker, in the hypothalamus. From day 15 following a transition to SD, most hypothalamic areas showed a decrease of PSA-NCAM level that was particularly significant in the preoptic area (POA). Following a transition to LD, PSA-NCAM content increased at day 15 in most regions except in the premammillary hypothalamic area (PMH) in which a significant decrease was noted. The functional importance of PSA-NCAM variations for seasonal reproduction was assessed for the PMH and POA. PSA-NCAM was degraded by stereotaxic injections of endoneuraminidase N and luteinising hormone (LH) secretion was recorded in treated and control ewes. Degradation of PSA-NCAM in the PMH in SD-treated ewes failed to produce a significant effect on LH secretion, whereas a similar treatment in the POA before a transition to SD delayed activation of the gonadotroph axis in two-thirds of the ewes. Our results suggest that the photoperiod controls variations of the hypothalamic content of a plasticity marker and that these might be important for the regulation of seasonal reproduction, particularly in the POA.


Assuntos
Hipotálamo/fisiologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Fotoperíodo , Reprodução/fisiologia , Ácidos Siálicos/fisiologia , Animais , Feminino , Hipotálamo/metabolismo , Melatonina/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Reprodução/efeitos dos fármacos , Ovinos , Ácidos Siálicos/metabolismo
19.
Alcohol Clin Exp Res ; 37(3): 383-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23050935

RESUMO

BACKGROUND: Fetal alcohol spectrum disorder is an immense public health problem. In vitro studies support the hypothesis that L1 cell adhesion molecule (L1) is a target for ethanol (EtOH) developmental neurotoxicity. L1 is critical for the development of the central nervous system. It functions through signal transduction leading to phosphorylation and dephosphorylation of tyrosines on its cytoplasmic domain. The function of L1 is also dependent on trafficking through lipid rafts (LRs). Our hypothesis is that L1 is a target for EtOH neurotoxicity in vivo. Our objective is to demonstrate changes in L1 phosphorylation/dephosphorylation and LR association in vivo. METHODS: Rat pups on postnatal day 6 are administered 4.5, 5.25, and 6 g/kg of EtOH divided into 2 doses 2 hours apart, then killed. Cerebella are rapidly frozen for assay. Blood is analyzed for blood EtOH concentration. L1 tyrosine phosphorylation is determined by immunoprecipitation and dephosphorylation of tyrosine 1176 determined by immunoblot. LRs are isolated by sucrose density gradient, and the distribution of L1 in LRs is determined. RESULTS: EtOH at all doses reduced the relative amount of Y1176 dephosphorylation as well as the relative amount of L1 phosphorylated on other tyrosines. The proportion of L1 present in LRs is significantly increased in pups who received 6 g/kg EtOH compared to intubated controls. CONCLUSIONS: L1 is a target for EtOH developmental neurotoxicity in vivo.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Etanol/administração & dosagem , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Regulação para Baixo/fisiologia , Feminino , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
20.
Neural Plast ; 2013: 805497, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691371

RESUMO

Depression is a devastating and prevalent disease, with profound effects on neural structure and function; however the etiology and neuropathology of depression remain poorly understood. Though antidepressant drugs exist, they are not ideal, as only a segment of patients are effectively treated, therapeutic onset is delayed, and the exact mechanism of these drugs remains to be elucidated. Several theories of depression do exist, including modulation of monoaminergic neurotransmission, alterations in neurotrophic factors, and the upregulation of adult hippocampal neurogenesis, and are briefly mentioned in the review. However none of these theories sufficiently explains the pathology and treatment of depression unto itself. Recently, neural plasticity theories of depression have postulated that multiple aspects of brain plasticity, beyond neurogenesis, may bridge the prevailing theories. The term "neural plasticity" encompasses an array of mechanisms, from the birth, survival, migration, and integration of new neurons to neurite outgrowth, synaptogenesis, and the modulation of mature synapses. This review critically assesses the role of adult hippocampal neurogenesis and the cell adhesion molecule, PSA-NCAM (which is known to be involved in many facets of neural plasticity), in depression and antidepressant treatment.


Assuntos
Transtorno Depressivo/fisiopatologia , Hipocampo/fisiopatologia , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Ácidos Siálicos/fisiologia , Animais , Transtorno Depressivo/metabolismo , Hipocampo/metabolismo , Humanos , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Ácidos Siálicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA