Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Endocrinol ; 190(1-2): 19-27, 2002 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11997175

RESUMO

The mammalian gonadotropin-releasing hormone receptor (GnRHR), with 327 amino acids, is among the smallest G protein coupled receptors identified. Absent from this receptor is the cytoplasmic tail, characteristic of other members of this superfamily, which frequently mediates desensitization and down-regulation. The fifteen carboxyl terminal residues in the mammalian GnRHR are absolutely conserved, suggesting important roles for these residues. In the current study, mutations of the mammalian GnRHR were made to study the carboxyl terminus. The receptor mutant GnRHR(Ser(326)Ala) was reduced in ligand affinity (117% reduction compared to wild type (wt)), while receptor numbers and internalization remained unchanged. GnRHR(Ser(326)Tyr) was decreased in effector coupling, while ligand affinity remained unchanged compared to wt. These studies also show that, while mutation of Ser(326) caused a change in ligand binding and effector coupling, truncation at this residue (GnRHR[des(326-327)]) had no measurable effect on GnRHR ligand binding, effector coupling or internalization, functions which appear to require different structural determinants than expression and routing. Removal of all three carboxyl terminal residues (Phe(325), Ser(326) and Leu(327)) or mutation of the receptor (GnRHR[Phe(325)Ala]) caused a complete loss of measurable ligand binding and effector coupling, clearly suggesting an unexplained role for Phe(325).


Assuntos
Endocitose/fisiologia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Ligantes , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Ligação Proteica , Ratos , Receptores LHRH/química
2.
Pediatr Res ; 61(6): 716-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17426653

RESUMO

Necrotizing enterocolitis (NEC) is a major cause of morbidity and death in premature infants. NEC is associated with increased levels of pro-inflammatory cytokines in plasma and tissues that are regulated by the transcription factor nuclear factor-kappaB (NF-kappaB). It remains unknown, however, whether NF-kappaB mediates injury in neonatal NEC. We therefore examined the activation status of NF-kappaB perinatally in the small intestine and in a neonatal rat model of NEC. We found that intestinal NF-kappaB is strongly activated at birth and, in dam-fed newborn rats, is down-regulated within a day. In contrast, NF-kappaB remains strongly activated at both d 1 and d 2 in stressed animals, and this is accompanied by a significant decrease in the levels of the endogenous NF-kappaB inhibitor protein IkappaBalpha and IkappaBbeta at d 2. To determine the importance of elevated NF-kappaB activity in intestinal injury in NEC, we administered the NEMO-binding domain (NBD) peptide that selectively inhibits the critical upstream IkappaB kinase (IKK). NBD but not a control peptide decreased mortality and bowel injury in this model, supporting the hypothesis that bowel injury in NEC results from elevated NF-kappaB activity. Our findings therefore lead us to conclude that selective NF-kappaB inhibition represents a promising therapeutic strategy for NEC.


Assuntos
Enterocolite Necrosante/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Peptídeos/uso terapêutico , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Óxido Nítrico Sintase Tipo II/análise , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley
3.
Immunology ; 118(2): 153-63, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16771850

RESUMO

CXCL2 (macrophage inflammatory protein-2 (MIP-2)), a critical chemokine for neutrophils, has been shown to be produced in the rat intestine in response to platelet-activating factor (PAF) and to mediate intestinal inflammation and injury. The intestinal epithelium, constantly exposed to bacterial products, is the first line of defence against micro-organisms. It has been reported that enterocytes produce proinflammatory mediators, including tumour necrosis factor (TNF) and PAF, and we showed that lipopolysaccharide (LPS) and TNF activate nuclear factor (NF)-kappaB in enterocytes. However, it remains elusive whether enterocytes release CXCL2 in response to LPS and TNF via a NF-kappaB-dependent pathway and whether this involves the endogenous production of TNF and PAF. In this study, we found that TNF and LPS markedly induced CXCL2 gene expression in IEC-6 cells, TNF within 30 min, peaking at 45 min, while LPS more slowly, peaking after 2 hr. TNF- and LPS- induced CXCL2 gene expression and protein release were completely blocked by pyrrolidine dithiocarbamate (PDTC) and helenalin, two potent NF-kappaB inhibitors. NEMO-binding domain peptide, a specific inhibitor of inhibitor protein kappaB kinase (IKK) activation, a major upstream kinase mediating NF-kappaB activation, significantly blocked CXCL2 gene expression and protein release induced by LPS. WEB2170 (PAF antagonist) and anti-TNF antibodies had no effect on LPS-induced CXCL2 expression. In conclusion, CXCL2 gene is expressed in enterocytes in response to both TNF and LPS. LPS-induced CXCL2 expression is dependent on NF-kappaB activation via the IKK pathway. The effect of LPS is independent of endogenous TNF and PAF.


Assuntos
Quimiocinas CXC/imunologia , Enterócitos/imunologia , Lipopolissacarídeos/imunologia , NF-kappa B/imunologia , Animais , Azepinas/farmacologia , Linhagem Celular , Quimiocina CXCL2 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Relação Dose-Resposta Imunológica , Ensaio de Desvio de Mobilidade Eletroforética , Enterócitos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/antagonistas & inibidores , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Fator de Ativação de Plaquetas/imunologia , Pirrolidinas/farmacologia , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transcrição Gênica/efeitos dos fármacos , Triazóis/farmacologia , Fator de Necrose Tumoral alfa/imunologia
4.
Am J Physiol Gastrointest Liver Physiol ; 287(6): G1220-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15319184

RESUMO

Platelet-activating factor (PAF) is a potent endogenous mediator of bowel inflammation. It activates neutrophils that are needed to initiate the inflammatory response. Macrophage inflammatory protein-2 (MIP-2), a critical C-X-C chemokine secreted by macrophages and epithelial cells, is a potent chemoattractant for neutrophils. Whereas MIP-2 has been previously shown to mediate the injury in various organs, its role in acute intestinal injury has never been assessed. In this study, we first investigated the effect of PAF on MIP-2 expression in the intestine. Anesthetized young adult male Sprague-Dawley rats were injected intravenously with either PAF (1.5 microg/kg) or saline. Sixty minutes later, ileal MIP-2 gene expression was determined by semiquantitative RT-PCR, and plasma and ileal MIP-2 protein was determined by ELISA. In a second step, we assessed the role of MIP-2 in PAF-induced bowel injury. Rats were pretreated with rabbit anti-rat MIP-2 antibodies or control IgG for 90 min and then injected intravenously with PAF (2.5 microg/kg) for 90 min. We found that, in the rat intestine, 1) MIP-2 mRNA was only minimally expressed constitutively in sham-operated animals; 2) MIP-2 mRNA was significantly upregulated in response to PAF; 3) MIP-2 protein plasma levels and local production of MIP-2 in the ileum were markedly induced by PAF; 4) the administration of anti-rat MIP-2 IgG, but not control rabbit IgG, markedly reduced PAF-induced bowel injury (injury scores of 0.19 +/- 0.09 vs. 1.12 +/- 0.43, P < 0.05), hypotension, and leukopenia but did not reduce PAF-induced hemoconcentration. Thus we conclude that MIP-2 mediates PAF-induced intestinal injury.


Assuntos
Mediadores da Inflamação/farmacologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/induzido quimicamente , Monocinas/biossíntese , Monocinas/genética , Fator de Ativação de Plaquetas/farmacologia , Animais , Anticorpos Bloqueadores/farmacologia , Quimiocina CXCL2 , Quimiocinas/farmacologia , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hematócrito , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Íleo/efeitos dos fármacos , Íleo/metabolismo , Íleo/patologia , Inflamação/induzido quimicamente , Mediadores da Inflamação/antagonistas & inibidores , Doenças Inflamatórias Intestinais/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Contagem de Leucócitos , Leucopenia/induzido quimicamente , Masculino , Monocinas/antagonistas & inibidores , Infiltração de Neutrófilos/efeitos dos fármacos , Peroxidase/biossíntese , Fator de Ativação de Plaquetas/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Immunology ; 106(4): 577-83, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12153521

RESUMO

Bacterial endotoxin (lipopolysaccharide; LPS) and platelet-activating factor (PAF) are important triggers of bowel inflammation and injury. We have previously shown that LPS activates the transcription factor nuclear factor (NF)-kappaB in the intestine, which up-regulates many pro-inflammatory genes. This effect partly depends on neutrophils and endogenous PAF. However, whether LPS and PAF directly activate NF-kappaB in enterocytes remains controversial. In this study, we first investigated the effect of LPS and PAF on NF-kappaB activation in IEC-6 (a non-transformed rat small intestinal crypt cell line) cells, by electrophoresis mobility shift assay and supershift, and found that LPS, but not PAF, activates NF-kappaB mostly as p50-p65 heterodimers. The effect was slower than tumour necrosis factor (TNF). Both LPS and TNF induce the expression of the NF-kappaB-dependent gene inducible nitric oxide synthase (iNOS), which occurs subsequent to NF-kappaB activation. We then examined the effect of LPS and TNF on the inhibitory molecules IkappaBalpha and IkappaBbeta. We found that TNF causes rapid degradation of IkappaBalpha and IkappaBbeta. In contrast, LPS did not change the levels of IkappaBalpha and IkappaBbeta up to 4 hr (by Western blot). However, in the presence of cycloheximide, there was a slow reduction of IkappaBalpha and IkappaBbeta, which disappeared almost completely at 4 hr. These observations suggest that LPS causes slow degradation and synthesis of IkappaBalpha and IkappaBbeta and therefore activates NF-kappaBeta via at least two mechanisms: initially, through an IkappaB-independent mechanism, and later, via an increased turnover of the inhibitor IkappaB. NF-kappaBeta activation precedes the gene expression of iNOS (assayed by reverse transcription-polymerase chain reaction), suggesting that LPS up-regulates iNOS via this transcription factor.


Assuntos
Endotoxinas/imunologia , Enterócitos/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Fator de Ativação de Plaquetas/imunologia , Animais , Western Blotting , Comunicação Celular/imunologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta Imunológica , Sinergismo Farmacológico , Enterócitos/imunologia , Leucócitos Mononucleares/imunologia , Lipopolissacarídeos/imunologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa