Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 172(1): 64-79, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25176008

RESUMO

BACKGROUND AND PURPOSE: Two peptide agonists of the glucagon-like peptide-1 (GLP-1) receptor, exenatide and GLP-1 itself, exert anti-hypersensitive effects in neuropathic, cancer and diabetic pain. In this study, we have assessed the anti-allodynic and anti-hyperalgesic effects of the non-peptide agonist WB4-24 in inflammatory nociception and the possible involvement of microglial ß-endorphin and pro-inflammatory cytokines. EXPERIMENTAL APPROACH: We used rat models of inflammatory nociception induced by formalin, carrageenan or complete Freund's adjuvant (CFA), to test mechanical allodynia and thermal hyperalgesia. Expression of ß-endorphin and pro-inflammatory cytokines was measured using real-time quantitative PCR and fluorescent immunoassays. KEY RESULTS: WB4-24 displaced the specific binding of exendin (9-39) in microglia. Single intrathecal injection of WB4-24 (0.3, 1, 3, 10, 30 and 100 µg) exerted dose-dependent, specific, anti-hypersensitive effects in acute and chronic inflammatory nociception induced by formalin, carrageenan and CFA, with a maximal inhibition of 60-80%. Spinal WB4-24 was not effective in altering nociceptive pain. Subcutaneous injection of WB4-24 was also antinociceptive in CFA-treated rats. WB4-24 evoked ß-endorphin release but did not inhibit expression of pro-inflammatory cytokines in either the spinal cord of CFA-treated rats or cultured microglia stimulated by LPS. WB4-24 anti-allodynia was prevented by a microglial inhibitor, ß-endorphin antiserum and a µ-opioid receptor antagonist. CONCLUSIONS AND IMPLICATIONS: Our results suggest that WB4-24 inhibits inflammatory nociception by releasing analgesic ß-endorphin rather than inhibiting the expression of proalgesic pro-inflammatory cytokines in spinal microglia, and that the spinal GLP-1 receptor is a potential target molecule for the treatment of pain hypersensitivity including inflammatory nociception.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios/farmacologia , Ciclobutanos/farmacologia , Microglia/efeitos dos fármacos , Receptores de Glucagon/agonistas , beta-Endorfina/metabolismo , Analgésicos/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Carragenina , Células Cultivadas , Ciclobutanos/uso terapêutico , Formaldeído , Adjuvante de Freund , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Temperatura Alta , Humanos , Peróxido de Hidrogênio , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Masculino , Microglia/metabolismo , Oxidantes , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Dor/induzido quimicamente , Dor/tratamento farmacológico , Dor/metabolismo , Ratos , Ratos Wistar , Coluna Vertebral/citologia
2.
Neuropharmacology ; 84: 31-45, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24747181

RESUMO

We recently discovered that the activation of the spinal glucagon-like peptide-1 receptors (GLP-1Rs) by the peptidic agonist exenatide produced antinociception in chronic pain. We suggested that the spinal GLP-1Rs are a potential target molecule for the management of chronic pain. This study evaluated the antinociceptive activities of geniposide, a presumed small molecule GLP-1R agonist. Geniposide produced concentration-dependent, complete protection against hydrogen peroxide-induced oxidative damage in PC12 and HEK293 cells expressing rat and human GLP-1Rs, but not in HEK293T cells that do not express GLP-1Rs. The orthosteric GLP-1R antagonist exendin(9-39) right-shifted the concentration-response curve of geniposide without changing the maximal protection, with identical pA2 values in both cell lines. Subcutaneous and oral geniposide dose-dependently blocked the formalin-induced tonic response but not the acute flinching response. Subcutaneous and oral geniposide had maximum inhibition of 72% and 68%, and ED50s of 13.1 and 52.7 mg/kg, respectively. Seven days of multidaily subcutaneous geniposide and exenatide injections did not induce antinociceptive tolerance. Intrathecal geniposide induced dose-dependent antinociception, which was completely prevented by spinal exendin(9-39), siRNA/GLP-1R and cyclic AMP/PKA pathway inhibitors. The geniposide iridoid analogs geniposidic acid, genipin methyl ether, 1,10-anhydrogenipin, loganin and catalpol effectively inhibited hydrogen peroxide-induced oxidative damage and formalin pain in an exendin(9-39)-reversible manner. Our results suggest that geniposide and its iridoid analogs produce antinociception during persistent pain by activating the spinal GLP-1Rs and that the iridoids represented by geniposide are orthosteric agonists of GLP-1Rs that function similarly in humans and rats and presumably act at the same binding site as exendin(9-39).


Assuntos
Analgésicos/administração & dosagem , Iridoides/administração & dosagem , Nociceptividade/efeitos dos fármacos , Receptores de Glucagon/metabolismo , Medula Espinal/efeitos dos fármacos , Analgésicos/química , Analgésicos/farmacologia , Animais , Fármacos do Sistema Nervoso Central/farmacologia , Exenatida , Formaldeído , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Compostos Heterocíclicos com 3 Anéis/química , Humanos , Glucosídeos Iridoides/administração & dosagem , Glucosídeos Iridoides/química , Iridoides/química , Iridoides/farmacologia , Masculino , Camundongos , Nociceptividade/fisiologia , Células PC12 , Fragmentos de Peptídeos/farmacologia , Peptídeos/administração & dosagem , Ratos , Ratos Wistar , Receptores de Glucagon/agonistas , Receptores de Glucagon/antagonistas & inibidores , Medula Espinal/fisiopatologia , Peçonhas/administração & dosagem
3.
J Neurosci ; 34(15): 5322-34, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24719110

RESUMO

This study aims to identify the inhibitory role of the spinal glucagon like peptide-1 receptor (GLP-1R) signaling in pain hypersensitivity and its mechanism of action in rats and mice. First, GLP-1Rs were identified to be specifically expressed on microglial cells in the spinal dorsal horn, and profoundly upregulated after peripheral nerve injury. In addition, intrathecal GLP-1R agonists GLP-1(7-36) and exenatide potently alleviated formalin-, peripheral nerve injury-, bone cancer-, and diabetes-induced hypersensitivity states by 60-90%, without affecting acute nociceptive responses. The antihypersensitive effects of exenatide and GLP-1 were completely prevented by GLP-1R antagonism and GLP-1R gene knockdown. Furthermore, exenatide evoked ß-endorphin release from both the spinal cord and cultured microglia. Exenatide antiallodynia was completely prevented by the microglial inhibitor minocycline, ß-endorphin antiserum, and opioid receptor antagonist naloxone. Our results illustrate a novel spinal dorsal horn microglial GLP-1R/ß-endorphin inhibitory pathway in a variety of pain hypersensitivity states.


Assuntos
Hiperalgesia/metabolismo , Neuralgia/metabolismo , Células do Corno Posterior/metabolismo , Receptores de Glucagon/agonistas , Animais , Células Cultivadas , Exenatida , Glucagon/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Humanos , Hiperalgesia/fisiopatologia , Microglia/metabolismo , Neuralgia/fisiopatologia , Nociceptividade , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Células do Corno Posterior/fisiologia , Ratos , Ratos Wistar , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Peçonhas/farmacologia , beta-Endorfina/metabolismo
4.
Br J Pharmacol ; 163(2): 399-412, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21244372

RESUMO

BACKGROUND AND PURPOSE: Exenatide is a 39-amino-acid peptide widely used to manage type 2 diabetes mellitus. However, it has a short plasma half-life and requires a twice daily injection regime. To overcome these drawbacks we used maleimide-polyethylene glycol to induce site-specific PEGylation. EXPERIMENTAL APPROACH: The analogue PB-105 (ExC39) was produced by replacing cysteine at position 39 of exenatide to provide a free thiol group. PB-105 showed the same glucoregulatory activity as exenatide in mice. Site-specific PEGylation of PB-105 was performed to produce PB-110 (ExC39PEG5kDa), PB-106 (ExC39PEG20kDa), PB-107 (ExC39PEG30kDa) and PB-108 (ExC39PEG40kDa). Their effects on intracellular cAMP, acute glucoregulatory activity and pharmacokinetic profile were compared in mice and rats. KEY RESULTS: PEGylation shifted the concentration-response curve of PB-105 to the right in a parallel, polyethylene glycol mass-dependent manner but with an inflexion point of at least 20 kDa. The activities of PB-107 and PB-108 but not PB-106 were reduced by 90% and 99%. PEGylation affected in vivo glucoregulatory activity in the same 'Inflexion-Shift' fashion at least at 20 kDa, but linearly increased plasma duration and systemic exposure without inflexion. PB-106 had a plasma t(1/2) approximately 10-fold that of PB-105, and exhibited superior glucoregulatory activity compared with PB-105 in normal and diabetic mice. CONCLUSIONS AND IMPLICATIONS: Site-specific PEGylation of exenatide with a permanent amide linkage affects its activity in a new type of 'Inflexion-Shift' fashion. PB-106 is a putative new analogue for treating diabetes; it possesses no loss of in vitro activity, prolonged plasma duration and superior, improved in vivo glucoregulatory activity compared with exenatide.


Assuntos
Hipoglicemiantes/farmacologia , Peptídeos/farmacologia , Polietilenoglicóis/química , Peçonhas/farmacologia , Sequência de Aminoácidos , Animais , Glicemia/análise , AMP Cíclico/biossíntese , Cisteína/química , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/tratamento farmacológico , Exenatida , Hipoglicemiantes/química , Hipoglicemiantes/farmacocinética , Masculino , Camundongos , Dados de Sequência Molecular , Células PC12 , Peptídeos/química , Peptídeos/farmacocinética , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Tirosina/química , Peçonhas/química , Peçonhas/farmacocinética
5.
Biochem Biophys Res Commun ; 404(1): 268-72, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21130077

RESUMO

DNA methylation and histone acetylation are major epigenetic modifications in gene silencing. In our previous research, we found that the methylated oligonucleotide (SurKex) complementary to a region of promoter of survivin could induce DNA methylation in a site-specific manner leading to survivin silencing. Here, we further studied the role of histone acetylation in survivin silencing and the relationship between histone acetylation and DNA methylation. First we observed the levels of histone H4 and H4K16 acetylation that were decreased after SurKex treatment by using the chromatin immunoprecipitation (ChIP) assay. Next, we investigated the roles of histone acetylation and DNA methylation in survivin silencing after blockade of histone deacetylation with Trichostatin A (TSA). We assessed survivin mRNA expression by RT-PCR, measured survivin promoter methylation by bisulfite sequencing and examined the level of histone acetylation by the ChIP assay. The results showed that histone deacetylation blocked by TSA reversed the effects of SurKex on inhibiting the expression of survivin mRNA, inducing a site-specific methylation on survivin promoter and decreasing the level of histone acetylation. Finally, we examined the role of histone acetylation in the expression of DNA methyltransferase 1 (DNMT1) mRNA. The results showed that histone deacetylation blocked by TSA reversed the increasing effect of histone deacetylation on the expression of survivin mRNA. This study suggests that histone deacetylation guides SurKex-induced DNA methylation in survivin silencing possibly through increasing the expression of DNMT1 mRNA.


Assuntos
Metilação de DNA , Inativação Gênica , Histonas/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Acetilação/efeitos dos fármacos , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas Inibidoras de Apoptose , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Survivina
6.
Biochem Biophys Res Commun ; 393(4): 592-7, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20152814

RESUMO

Anti-apoptotic survivin is over-expressed in a variety of human carcinomas and is considered as a therapeutic target in cancers. Suppression of survivin mRNA by RNAi and anti-sense nucleotides has proved to be a powerful anti-tumor therapy in both animal models and human investigations. In this communication, we tested an alternative approach to silence survivin by knocking down its gene transcription through a short methylated oligonucleotide (SurKex) that is complementary to the survivin gene promoter. Treatment of NCI-H460 cells with SurKex significantly suppressed the production of survivin mRNA and its oncoprotein. DNA bisulfite sequencing showed that SurKex induced site-specific de novo CpG methylation in the complementary region of survivin promoter. Chromatin immunoprecipitation assay also demonstrated that SurKex induced histone hypermethylation at histone H3K9 and H3K27 as well as deacetylation of histone H4 in the same regulatory region. SurKex remarkably inhibited tumor growth in nude mice bearing xenograft tumors. This study demonstrates that the synthetic methylated oligonucleotide SurKex inhibits tumor growth by silencing the survivin gene using a mechanism of altering the epigenotype in the survivin promoter. Thus, targeted epigenetic modification in the gene promoter may offer a new general strategy to silence tumor-related genes in tumor therapy.


Assuntos
Cromatina/metabolismo , Metilação de DNA , Inativação Gênica , Neoplasias Pulmonares/terapia , Proteínas Associadas aos Microtúbulos/genética , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos/uso terapêutico , Animais , Sequência de Bases , Linhagem Celular Tumoral , Cromatina/genética , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Epigênese Genética , Humanos , Proteínas Inibidoras de Apoptose , Neoplasias Pulmonares/genética , Camundongos , Dados de Sequência Molecular , Oligonucleotídeos/genética , Oligonucleotídeos Antissenso/genética , RNA Interferente Pequeno/genética , Proteínas Repressoras , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA