Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 110(14): 5683-8, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23431142

RESUMO

There is a genetic contribution to fetal alcohol spectrum disorders (FASD), but the identification of candidate genes has been elusive. Ethanol may cause FASD in part by decreasing the adhesion of the developmentally critical L1 cell adhesion molecule through interactions with an alcohol binding pocket on the extracellular domain. Pharmacologic inhibition or genetic knockdown of ERK2 did not alter L1 adhesion, but markedly decreased ethanol inhibition of L1 adhesion in NIH/3T3 cells and NG108-15 cells. Likewise, leucine replacement of S1248, an ERK2 substrate on the L1 cytoplasmic domain, did not decrease L1 adhesion, but abolished ethanol inhibition of L1 adhesion. Stable transfection of NIH/3T3 cells with human L1 resulted in clonal cell lines in which L1 adhesion was consistently sensitive or insensitive to ethanol for more than a decade. ERK2 activity and S1248 phosphorylation were greater in ethanol-sensitive NIH/3T3 clonal cell lines than in their ethanol-insensitive counterparts. Ethanol-insensitive cells became ethanol sensitive after increasing ERK2 activity by transfection with a constitutively active MAP kinase kinase 1. Finally, embryos from two substrains of C57BL mice that differ in susceptibility to ethanol teratogenesis showed corresponding differences in MAPK activity. Our data suggest that ERK2 phosphorylation of S1248 modulates ethanol inhibition of L1 adhesion by inside-out signaling and that differential regulation of ERK2 signaling might contribute to genetic susceptibility to FASD. Moreover, identification of a specific locus that regulates ethanol sensitivity, but not L1 function, might facilitate the rational design of drugs that block ethanol neurotoxicity.


Assuntos
Adesão Celular/efeitos dos fármacos , Etanol/toxicidade , Transtornos do Espectro Alcoólico Fetal/fisiopatologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Análise de Variância , Animais , Feminino , Transtornos do Espectro Alcoólico Fetal/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/genética , Células NIH 3T3 , Molécula L1 de Adesão de Célula Nervosa/genética , Fosforilação , Gravidez
2.
Proc Natl Acad Sci U S A ; 105(1): 371-5, 2008 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18165316

RESUMO

Prenatal ethanol exposure causes fetal alcohol spectrum disorders (FASD) in part by disrupting the neural cell adhesion molecule L1. L1 gene mutations cause neuropathological abnormalities similar to those of FASD. Ethanol and 1-butanol inhibit L1-mediated cell-cell adhesion (L1 adhesion), whereas 1-octanol antagonizes this action. To test the hypothesis that there are alcohol binding sites on L1, we used 3-azibutanol and 3-azioctanol, the photoactivatable analogs of 1-butanol and 1-octanol, to photolabel the purified Ig1-4 domain of human L1 (hL1 Ig1-4). 3-Azibutanol (11 mM), like ethanol, inhibited L1 adhesion in NIH/3T3 cells stably transfected with hL1, whereas subanesthetic concentrations of 3-azioctanol (14 microM) antagonized ethanol inhibition of L1 adhesion. 3-Azibutanol (100-1,000 microM) and 3-azioctanol (10-100 microM) photoincorporated into Tyr-418 on Ig4 and into two adjacent regions in the N terminus, Glu-33 and Glu-24 to Glu-27. A homology model of hL1 Ig1-4 (residues 33-422), based on the structure of the Ig1-4 domains of axonin-1, suggests that Glu-33 and Tyr-418 hydrogen-bond at the interface of Ig1 and Ig4 to stabilize a horseshoe conformation of L1 that favors homophilic binding. Furthermore, this alcohol binding pocket lies within 7 A of Leu-120 and Gly-121, residues in which missense mutations cause neurological disorders similar to FASD. These data suggest that ethanol or selected mutations produce neuropathological abnormalities by disrupting the domain interface between Ig1 and Ig4. Characterization of alcohol agonist and antagonist binding sites on L1 will aid in understanding the molecular basis for FASD and might accelerate the development of ethanol antagonists.


Assuntos
Álcoois/química , Butanóis/farmacologia , Moléculas de Adesão Celular/metabolismo , Diazometano/análogos & derivados , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Animais , Sítios de Ligação , Adesão Celular , Diazometano/farmacologia , Etanol/química , Ligação de Hidrogênio , Luz , Espectrometria de Massas/métodos , Camundongos , Modelos Biológicos , Modelos Genéticos , Conformação Molecular , Células NIH 3T3 , Molécula L1 de Adesão de Célula Nervosa/química , Fotoquímica/métodos
3.
Dev Neurosci ; 27(1): 13-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15886480

RESUMO

Ethanol inhibition of L1-mediated cell adhesion may contribute to the spectrum of neurological, behavioral and morphological abnormalities associated with prenatal ethanol exposure. We showed previously that the neuroprotective peptides NAPVSIPQ (NAP) and SALLRSIPA (SAL) antagonize ethanol inhibition of L1 adhesion and prevent ethanol-induced growth retardation in mouse whole embryo culture. Here we ask whether NAP and SAL also prevent ethanol-induced major malformations of the nervous system. Gestational day 8.0 (3-5 somites) C57BL/6J mouse embryos were grown for 6 h in control medium, 100 mM ethanol and 10(-10) M peptides and then maintained for an additional 20 h in control medium. At the end of the culture period, only embryos having 18-19 somite pairs were examined and compared for the degree of neural tube closure. Ethanol exposure resulted in neural tube defects (NTDs) consistent with total dysraphia and anencephaly. Co-incubation with ethanol and L-NAP (all L-amino acids), D-NAP (all D-amino acids) or SAL significantly increased the percentage of embryos that had begun to close their neural folds at the level of the forebrain/midbrain junction or that had progressed beyond this stage of closure. P7A-NAP (NAPVSIAQ), which lacks neuroprotective activity, but retains activity as an antagonist of ethanol inhibition of L1 adhesion, was effective in preventing ethanol-induced NTDs. In contrast, I6A-NAP (NAPVSAPQ), which shows reduced efficacy as an ethanol antagonist but retains its neuroprotective efficacy, did not significantly diminish the induction of NTDs by ethanol. These findings demonstrate the ability of NAP and SAL to prevent ethanol-induced NTDs and support the hypothesis that ethanol teratogenesis is caused in part by ethanol inhibition of L1-mediated cell adhesion.


Assuntos
Etanol/toxicidade , Defeitos do Tubo Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Animais , Encéfalo/anormalidades , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Modelos Animais de Doenças , Técnicas de Cultura Embrionária , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/patologia , Oligopeptídeos/farmacologia , Peptídeos/química , Gravidez
4.
J Pharmacol Exp Ther ; 309(3): 1183-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14762101

RESUMO

Increasing evidence suggests that ethanol damages the developing nervous system partly by disrupting the L1 cell adhesion molecule. Ethanol inhibits L1-mediated cell adhesion, and compounds that antagonize this action also prevent ethanol-induced embryotoxicity. Two such compounds are the small peptides NAPVSIPQ (NAP) and SALLRSIPA (SAL). We showed previously that NAP and SAL antagonize ethanol inhibition of L1 adhesion at femtomolar to picomolar concentrations. Here we demonstrate that, despite this extraordinary potency, both NAP and SAL lack stereospecificity. d-NAP, a peptide composed entirely of d-amino acids, was an effective ethanol antagonist in NIH/3T3 cells transfected with human L1 and in the NG108-15 neural cell line. Interestingly, Ala-substituted derivatives of d-NAP demonstrate the same structure-activity relation as the corresponding derivatives of l-NAP. The Ser-Ile-Pro motif was important for the ethanol antagonist activity of d-NAP, l-NAP, and l-SAL, with Ile being the most critical element in all three. Like l-NAP, d-NAP effectively reduced ethanol-induced growth retardation in mouse whole embryo culture. The potential resistance of d-peptides to proteases makes d-NAP a potentially attractive agent for the prevention of fetal alcohol syndrome.


Assuntos
Etanol/antagonistas & inibidores , Oligopeptídeos/farmacologia , Células 3T3 , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Embrião de Mamíferos/efeitos dos fármacos , Etanol/toxicidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/química , Peptídeos/farmacologia , Relação Estrutura-Atividade
5.
Proc Natl Acad Sci U S A ; 100(14): 8543-8, 2003 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-12808140

RESUMO

NAPVSIPQ (NAP), an active fragment of the glial-derived activity-dependent neuroprotective protein, is protective at femtomolar concentrations against a wide array of neural insults and prevents ethanol-induced fetal wastage and growth retardation in mice. NAP also antagonizes ethanol inhibition of L1-mediated cell adhesion (ethanol antagonism). We performed an Ala scanning substitution of NAP to determine the role of ethanol antagonism and neuroprotection in NAP prevention of ethanol embryotoxicity. The Ser-Ile-Pro region of NAP was crucial for both ethanol antagonism and protection of cortical neurons from tetrodotoxin toxicity (neuroprotection). Ala replacement of either Ser-5 or Pro-7 (P7A-NAP) abolished NAP neuroprotection but minimally changed the efficacy of NAP ethanol antagonism. In contrast, Ala replacement of Ile-6 (I6A-NAP) caused a decrease in potency (>2 logarithmic orders) with only a small reduction (<10%) in the efficacy of NAP neuroprotection but markedly reduced the efficacy (50%) and the potency (5 logarithmic orders) of NAP ethanol antagonism. Ethanol significantly reduced the number of paired somites in mouse whole-embryo culture; this effect was prevented significantly by 100 pM NAP or by 100 pM P7A-NAP, but not by 100 pM I6A-NAP. The structure-activity relation for NAP prevention of ethanol embryotoxicity was similar to that for NAP ethanol antagonism and different from that for NAP neuroprotection. These findings support the hypothesis that NAP antagonism of ethanol inhibition of L1 adhesion plays a central role in NAP prevention of ethanol embryotoxicity and highlight the potential importance of ethanol effects on L1 in the pathophysiology of fetal alcohol syndrome.


Assuntos
Anormalidades Induzidas por Medicamentos/prevenção & controle , Etanol/antagonistas & inibidores , Proteínas de Homeodomínio , Proteínas do Tecido Nervoso/química , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Anormalidades Induzidas por Medicamentos/embriologia , Substituição de Aminoácidos , Animais , Adesão Celular/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Etanol/toxicidade , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/química , Oligopeptídeos/química , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/química , Gravidez , Relação Estrutura-Atividade , Tetrodotoxina/toxicidade
6.
Mol Pharmacol ; 62(5): 1053-60, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12391267

RESUMO

1-Octanol antagonizes ethanol inhibition of L1-mediated cell adhesion and prevents ethanol teratogenesis in mouse whole embryo culture. Herein, we identify a new series of alcohol antagonists and study their mechanism of action. Cell aggregation assays were carried out in ethanol-sensitive, human L1-transfected NIH/3T3 cells in the absence and presence of 100 mM ethanol or 2 mM 1-butanol and candidate antagonists. Antagonist potency for 1-alcohols increased progressively over 5 log orders from 1-pentanol (C5) to 1-dodecanol (C12). Antagonist potency declined from 1-dodecanol (C12) to 1-tridecanol (C13), and 1-tetradecanol (C14) and 1-pentadecanol (C15) were inactive. The presence and position of a double bond in the 1-butanol molecule determined whether a compound was a full agonist (1-butanol), a mixed agonist-antagonist (2-buten-1-ol), or an antagonist (3-buten-1-ol). Increasing the concentration of agonist (1-butanol or ethanol) overcame the antagonism of 3-buten-1-ol, benzyl alcohol, cyclopentanol, and 3-pentanol, but not that of 4-methyl-1-pentanol, 2-methyl-2-pentanol, 1-pentanol, 2-pentanol, 1-octanol, and 2,6-di-isopropylphenol (propofol), suggesting that the mechanisms of antagonism may differ between these groups of compounds. These findings suggest that selective straight, branched, and cyclic alcohols may act at multiple, discrete sites to antagonize the actions of ethanol and 1-butanol on L1-mediated cell-cell adhesion.


Assuntos
1-Octanol/farmacologia , Adesão Celular/efeitos dos fármacos , Etanol/farmacologia , 1-Butanol/química , 1-Butanol/farmacologia , 1-Octanol/química , Células 3T3 , Animais , Adesão Celular/fisiologia , Etanol/química , Camundongos , Relação Estrutura-Atividade
7.
J Pharmacol Exp Ther ; 303(1): 110-6, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12235240

RESUMO

Ethanol inhibits cell-cell adhesion mediated by the L1 cell adhesion molecule. 1-Octanol potently antagonizes this cellular action of ethanol and also prevents ethanol-induced dysmorphology and cell death in mouse whole embryo culture. NAPVSIPQ (NAP) and SALLRSIPA (SAL) are active peptide fragments of two neuroprotective proteins: activity-dependent neuroprotective protein and activity-dependent neurotrophic factor. NAP and SAL are neuroprotective at femtomolar concentrations against a variety of neurotoxins and also prevent ethanol teratogenesis in mice. To explore the cellular basis for this action, we asked whether NAP and SAL antagonize ethanol inhibition of L1 adhesion. Aggregation assays were carried out in ethanol-sensitive, human L1-transfected NIH/3T3 cells in the absence and presence of NAP and SAL. Neither NAP nor SAL altered L1 adhesion or L1 expression; however, both peptides potently and completely antagonized the inhibition of L1 adhesion by 100 mM ethanol (EC(50): NAP, 6 x 10(-14) M; SAL, 4 x 10(-11) M). NAP also antagonized ethanol inhibition of cell-cell adhesion in bone morphogenetic protein-7-treated NG108-15 cells. In L1-expressing NIH/3T3 cells, SAL antagonism was reversible and could be overcome by increasing concentrations of ethanol. In contrast, NAP antagonism was irreversible and could not be overcome by increasing agonist concentration. Two scrambled NAP peptides (ASPNQPIV and PNIQVASP) were not antagonists at concentrations as high as 10(-7) M. Thus, two structurally unrelated classes of compounds, alcohols and small polypeptides, share two common actions: antagonism of ethanol inhibition of L1-mediated cell adhesion and prevention of ethanol teratogenesis. These findings support the hypothesis that ethanol inhibition of L1 adhesion contributes to ethanol teratogenesis.


Assuntos
Adesão Celular/efeitos dos fármacos , Etanol/farmacologia , Oligopeptídeos/farmacologia , Células 3T3 , Sequência de Aminoácidos , Animais , Adesão Celular/fisiologia , Linhagem Celular , Etanol/antagonistas & inibidores , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...