Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuropeptides ; 97: 102309, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36410163

RESUMO

The chimeric peptide EN-9 was reported as a κ-opioid/neuropeptide FF receptors bifunctional agonist that modulated chronic pain with no tolerance. Many lines of evidence have shown that the effect of the κ-opioid receptor is mediated by not only its specific activation but also downstream events participation, especially interaction with the µ-opioid receptor pathway in antinociception and tolerance on most occasions. The present study investigated the acute and chronic cross-tolerance of EN-9 with µ-opioid receptor agonist EM-2, DAMGO, and morphine after intracerebroventricularly (i.c.v) injection in the mouse tail-flick test. In the acute tolerance test, EN-9 showed symmetrical acute cross-tolerance to DAMGO but no cross-tolerance to EM2. In the chronic tolerance test, EN-9 had no tolerance after eight days of repeated administration. However, EN-9 illustrated complete cross-tolerance to morphine and symmetrical cross-tolerance to EM2. In addition, inhibition of NPFF receptor could induce the tolerance development of EN-9. These findings indicated that supraspinal EN-9-induced antinociception contains additional components, which are mediated by the downstream µ-opioid receptor pathway both in acute and chronic treatment, whereas the subtypes of µ-opioid receptor or NPFF system pathway involved in antinociceptive effects induced by EN-9 are complex. Identifying the receptor mechanism could help design preferable bifunctional opioid compounds.


Assuntos
Analgésicos Opioides , Analgésicos , Camundongos , Animais , Analgésicos Opioides/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Analgésicos/farmacologia , Receptores Opioides mu/metabolismo , Morfina/farmacologia
2.
Aging (Albany NY) ; 13(4): 5087-5103, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33535180

RESUMO

This study aimed to investigate the therapeutic mechanism of Huankuile suspension (HKL), a typical traditional Chinese medicine, on ulcerative colitis (UC) in a rat model. UC model was established by 2,4,6-trinitrobenzene sulfonic acid (TNBS) enema. Then, the rats were randomly divided into three groups: water treated group, HKL treated group and 5- amino salicylic acid (5-ASA) treated group. After 7 days treatment, the histological score in the HKL treated group was comparable with those in the control group. qRT-PCR and western blot demonstrated that HKL could significantly decreased pro-inflammatory cytokines, including TNF-α, IL-1ß and IL-6, while having less effect on anti-inflammatory cytokines, including IL-4 and IL-10. Transcriptomic analysis identified 670 differentially expressed genes (DEGs) between HKL treated UC rats and water treated UC rats. These DEGs were mostly related with immune response. Besides, metabonomic profile revealed 136 differential metabolites which were significantly enriched in "pyrimidine metabolism", "glutathione metabolism", "purine metabolism" and "citrate cycle". Finally, integrated analysis revealed that metabonomic pathways including "steroid hormone biosynthesis", "pyrimidine metabolism", "purine metabolism", and "glutathione metabolism" were altered by HKL at both transcriptomic and metabonomic levels. HKL could inhibit inflammation and regulate bile metabolism, pyrimidine metabolism, purine metabolism, glutathione metabolism and citrate cycle.


Assuntos
Colite Ulcerativa/genética , Citocinas/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/patologia , Colite Ulcerativa/fisiopatologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mesalamina/farmacologia , Metabolômica , Ratos , Ácido Trinitrobenzenossulfônico , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
Bioorg Med Chem ; 27(4): 630-643, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626554

RESUMO

It is well known that opioid analgesics produce side effects including tolerance and constipation. Since neuropeptide FF (NPFF) receptor antagonists reversed opioid-induced hyperalgesia and analgesic tolerance, the present work was performed to synthetize two branched peptidomimetics, EKR and RKE, containing the opioid peptide endomorphin-2 (EM-2) and the NPFF receptor antagonist RF9. Our data obtained from the in vitro cyclic adenosine monophosphate experiment demonstrated that EKR functioned as a mixed mu-, delta-opioid receptors agonist and NPFF1 receptor antagonist/NPFF2 receptor partial agonist, whereas RKE acted as a multi-functional peptidomimetic with the mu-opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism. Furthermore, EKR and RKE completely blocked the NPFF2 receptor-mediated neurite outgrowth of Neuro 2A cells. In vivo antinociception studies found that supraspinal administration of EKR and RKE dose-dependently produced potent antinociception via the mu-opioid receptor in the tail-flick test. In carrageenan inflammatory pain model, spinal administration of EKR and RKE induced dose-related analgesia, which was significantly reduced by the opioid antagonist naloxone and the NPFF antagonist RF9. Notably, compared with morphine, intracerebroventricular repeated administration of EKR and RKE maintained prolonged antinociceptive effectiveness. In addition, at the antinociceptive doses, these two branched peptidomimetics did not significantly inhibit gastrointestinal transit. Taken together, the present work suggest that EKR and RKE behave as multi-functional ligands with the opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism, and produce prolonged antinociception with limited side effects. Moreover, our results imply that EKR and RKE might be interesting pharmacological tools for further investigating the biological function of the NPFF and opioid systems.


Assuntos
Analgésicos Opioides/farmacologia , Descoberta de Drogas , Peptidomiméticos/farmacologia , Analgésicos Opioides/síntese química , Analgésicos Opioides/uso terapêutico , Animais , Linhagem Celular Tumoral , Agonismo Parcial de Drogas , Trânsito Gastrointestinal/efeitos dos fármacos , Células HEK293 , Humanos , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Morfina/farmacologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Peptidomiméticos/síntese química , Peptidomiméticos/uso terapêutico , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
4.
Eur J Pharmacol ; 837: 53-63, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30172787

RESUMO

We recently characterized a novel bifunctional agonist for opioid and neuropeptide FF receptors, named BN-9, which exhibited potent analgesia in the mouse tail-flick test when given centrally. To further evaluate its potential therapeutic efficacy for translational-medical development, the current work was performed to explore the antinociceptive activities of intraperitoneal (i.p.) administration of BN-9 in mouse models of tail-flick assay, formalin pain, visceral pain and post-operative pain. In the tail-flick test, BN-9 induced a dose-related antinociceptive effect, which was fully blocked by systemic pretreatment with the peripheral acting opioid receptor antagonist naloxone methiodide, but not supraspinal naloxone methiodide, implying the involvement of the peripheral opioid receptors. In addition, the systemic antinociception of BN-9 was antagonized by the selective antagonists of the µ- and κ-opioid receptors, independently of the δ-opioid and neuropeptide FF receptors. Similarly, dose-dependent analgesia was also produced by systemic BN-9 in different pain models via the peripheral opioid receptors, independently of the neuropeptide FF receptors. Furthermore, the side-effects of systemic BN-9 on motor performance, tolerance development and gastrointestinal transit inhibition were also evaluated. Repeated systemic injection of BN-9 produced non-tolerance analgesia over 8 days. Compared with morphine, intraperitoneal administration of BN-9 exerted less inhibition of gastrointestinal transit. These data show that BN-9 induced systemic analgesia with reduced side-effects on tolerance and constipation. This article suggests that systemic injection of BN-9 causes effective antinociception in different preclinical pain models via the peripheral opioid receptors, providing an attractive approach to develop peripherally acting opioid analgesics with multiple targeting properties.


Assuntos
Analgésicos/uso terapêutico , Oligopeptídeos/uso terapêutico , Dor/tratamento farmacológico , Receptores de Neuropeptídeos/agonistas , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Tolerância a Medicamentos , Trânsito Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacologia
5.
Parasitol Res ; 116(5): 1573-1579, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28361273

RESUMO

To date, little is known about cytosine methylation in the genomic DNA of apicomplexan parasites, although it has been confirmed that this important epigenetic modification exists in many lower eukaryotes, plants, and animals. In the present study, ELISA-based detection demonstrated that low levels of 5-methylcytosine (5-mC) are present in Eimeria spp., Toxoplasma gondii, Cryptosporidium spp., and Neospora caninum. The proportions of 5-mC in genomic DNA were 0.18 ± 0.02% in E tenella sporulated oocysts, 0.19 ± 0.01% in E. tenella second-generation merozoites, 0.22 ± 0.04% in T. gondii tachyzoites, 0.28 ± 0.03% in N. caninum tachyzoites, and 0.06 ± 0.01, 0.11 ± 0.01, and 0.09 ± 0.01% in C. andersoni, C. baileyi, and C. parvum sporulated oocysts, respectively. In addition, we found that the percentages of 5-mC in E. tenella varied considerably at different life stages, with sporozoites having the highest percentage of 5-mC (0.78 ± 0.10%). Similar stage differences in 5-mC were also found in E. maxima, E. necatrix, and E. acervulina, the levels of 5-mC in their sporozoites being 4.3-, 1.8-, 2.5-, and 2.0-fold higher than that of sporulated oocysts, respectively (p < 0.01). Furthermore, a total DNA methyltransferase-like activity was detected in whole cell extracts prepared from E. tenella sporozoites. In conclusion, genomic DNA methylation is present in these apicomplexan parasites and may play a role in the stage conversion of Eimeria.


Assuntos
5-Metilcitosina/análise , Galinhas/parasitologia , Coccidiose/veterinária , Cryptosporidium/genética , DNA de Protozoário/genética , Eimeria tenella/genética , Neospora/genética , Toxoplasma/genética , Animais , Coccidiose/parasitologia , Metilação de DNA/genética , Ensaio de Imunoadsorção Enzimática , Merozoítos/citologia , Oocistos/citologia , Esporozoítos/citologia
6.
Neuropeptides ; 63: 83-90, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28010996

RESUMO

Hemopressin and related peptides have shown to function as the endogenous ligands or the regulator of cannabinoid receptors. Moreover, hemopressin and its truncated peptides were also reported to produce a slight modulatory effect on opioid system. In the present work, based on the amino acid sequence analyses of hemoglobin subunit α, rat VD-hemopressin(α) [(r)VD-Hpα] was predicted as a cannabinoid peptide derived from rat α-hemoglobin. Furthermore, (r)VD-Hpα was synthesized and characterized in a series of in vitro and in vivo assays. Our results demonstrated that (r)VD-Hpα induced neurite outgrowth in Neuro 2A cells via CB1 receptor. In the tail-flick assay, (r)VD-Hpα dose-dependently exerted central antinociception through CB1 receptor, but not CB2 and opioid receptors. In mice, supraspinal administration of (r)VD-Hpα produced dose-dependent hypothermia, which was partially reduced by the CB1 receptor antagonist AM251, but not by the antagonists of CB2 and opioid receptors. In addition, (r)VD-Hpα caused hypoactivity after intracerebroventricular injection, and this effect was insensitive to the antagonists of cannabinoid and opioid receptors. Further assessment of the side-effects demonstrated that (r)VD-Hpα evoked the limited effects on gastrointestinal transit at antinociceptive doses, but repeated i.c.v. injection of (r)VD-Hpα induced development of antinociceptive tolerance. Taken together, these data suggest that the predicted peptide (r)VD-Hpα produces antinociception, hypothermia and hypoactivity via different pharmacological mechanisms, at least partially, which may offer an attractive strategy for separating cannabinoid analgesia from hypoactivity. Moreover, it implies that (r)VD-Hpα has therapeutic potential in pain management with limited side-effects.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Oligopeptídeos/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Trânsito Gastrointestinal/efeitos dos fármacos , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Crescimento Neuronal/fisiologia , Neurônios/metabolismo , Nociceptividade/efeitos dos fármacos , Medição da Dor
7.
Br J Pharmacol ; 173(11): 1864-80, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27018797

RESUMO

BACKGROUND AND PURPOSE: Neuropeptide FF (NPFF) behaves as an endogenous opioid-modulating peptide. In the present study, the opioid and NPFF pharmacophore-containing chimeric peptide BN-9 was synthesized and pharmacologically characterized. EXPERIMENTAL APPROACH: Agonist activities of BN-9 at opioid and NPFF receptors were characterized in in vitro cAMP assays. Antinociceptive activities of BN-9 were evaluated in the mouse tail-flick and formalin tests. Furthermore, its side effects were investigated in rotarod, antinociceptive tolerance, reward and gastrointestinal transit tests. KEY RESULTS: BN-9 acted as a novel multifunctional agonist at µ, δ, κ, NPFF1 and NPFF2 receptors in cAMP assays. In the tail-flick test, BN-9 produced dose-related antinociception and was approximately equipotent to morphine; this antinociception was blocked by µ and κ receptor antagonists, but not by the δ receptor antagonist. In the formalin test, supraspinal administration of BN-9 produced significant analgesia. Notably, repeated administration of BN-9 produced analgesia without loss of potency over 8 days. In contrast, repeated i.c.v. co-administration of BN-9 with the NPFF receptor antagonist RF9 produced significant antinociceptive tolerance. Furthermore, i.c.v. BN-9 induced conditioned place preference. When given by the same routes, BN-9 had a more than eightfold higher ED50 value for gastrointestinal transit inhibition compared with the ED50 values for antinociception. CONCLUSIONS AND IMPLICATIONS: BN-9 produced a robust, nontolerance-forming analgesia with limited inhibition of gastrointestinal transit. As BN-9 is able to activate both opioid and NPFF systems, this provides an interesting approach for the development of novel analgesics with minimal side effects.


Assuntos
Analgésicos/farmacologia , Oligopeptídeos/farmacologia , Dor/tratamento farmacológico , Receptores de Neuropeptídeos/agonistas , Receptores Opioides/agonistas , Analgésicos/efeitos adversos , Analgésicos/química , Animais , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Relação Estrutura-Atividade
8.
Neuropharmacology ; 108: 364-72, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26970017

RESUMO

Mounting evidences indicate the functional interactions between neuropeptide FF (NPFF) and opioids, including the endogenous opioids. In the present work, EN-9, a chimeric peptide containing the functional domains of the endogenous opioid endomorphin-2 (EM-2) and NPFF, was synthesized and pharmacologically characterized. In vitro cAMP assay demonstrated that EN-9 was a multifunctional agonist of κ-opioid, NPFF1 and NPFF2 receptors. In the mouse tail-flick test, intracerebroventricularly (i.c.v.) administration of EN-9 produced significant antinociception with an ED50 value of 13.44 nmol, which lasted longer than that of EM-2. In addition, EN-9 induced potent antinociception after both intravenous (i.v.) and subcutaneous (s.c.) injection. Furthermore, the experiments using the antagonists of opioid and NPFF receptors indicated that the central antinociception of EN-9 was mainly mediated by κ-opioid receptor, independently on NPFF receptors. Notably, the central antinociception of EN-9 was not reduced over a period of 6 days repeated i.c.v. injection. Repeated i.c.v. administration of EN-9 with the NPFF1 and NPFF2 receptors antagonist RF9 resulted in a progressive loss of analgesic potency, consistent with the development of tolerance. Moreover, central administration of EN-9 induced the place conditioning aversion only at a high dose of 60 nmol, but not at low doses. At supraspinal level, only high dose of EN-9 (60 nmol, i.c.v.) inhibited gastrointestinal transit via NPFF receptors. Similarly, systemic administration of EN-9 also inhibited gastrointestinal transit at high doses (10 and 30 mg/kg, i.v.). Taken together, the multifunctional agonist of κ-opioid and NPFF receptors EN-9 produced a potent, non-tolerance forming antinociception with limited side effects.


Assuntos
Analgésicos Opioides/administração & dosagem , Oligopeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Analgésicos/administração & dosagem , Analgésicos/química , Analgésicos Opioides/química , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/fisiologia , Células HEK293 , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Oligopeptídeos/química , Medição da Dor/métodos , Fragmentos de Peptídeos/química , Peptídeos/química , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/fisiologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/fisiologia
9.
Anesth Analg ; 121(5): 1360-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26273748

RESUMO

BACKGROUND: Cannabinoids produce analgesia in several pain models, but the undesirable side effects from high doses of cannabinoid drugs limit their clinic use. Our recent results indicate that cannabinoid-induced antinociception was enhanced by neuropeptide VF (NPVF). Here, we investigate whether low-dose cannabinoid agonists combined with NPVF can produce effective antinociception with limited side effects. METHODS: The in vivo properties of (R)-(-1)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-napthalenylmethanone (WIN55,212-2) given alone and its combination with NPVF were evaluated in nociceptive modulation, locomotor activity, gastrointestinal transit, and tolerance development assays after intracerebroventricular administration in mice. RESULTS: In the radiant tail-flick test, the antinociception of combination of WIN55,212-2 and NPVF was more potent than that of cannabinoid agonist given alone, with an ED50 shift from 3.51 to 0.69 nmol; 9 nmol WIN55,212-2 alone and 3 nmol WIN55,212-2 combined with NPVF induced equivalent antinociception after supraspinal administration. The cannabinoid-potentiating effects of NPVF were reduced by both the cannabinoid receptor type 1 and the neuropeptide FF receptor antagonists. In the formalin assay, WIN55,212-2 combined with NPVF also significantly reduced pain-related behaviors. However, the combination of WIN55,212-2 with NPVF exerted significant hypoactivity in a manner similar to high doses of WIN55,212-2. It was important to note that the combination of WIN55,212-2 with NPVF produced nontolerance-forming antinociception and weaker inhibition of gastrointestinal transit compared with high dose of WIN55,212-2. CONCLUSIONS: These data suggest that the cannabinoid agonist combined with NPVF produces effective antinociception-lacking tolerance via both cannabinoid receptor type 1 and neuropeptide FF receptors in the brain.


Assuntos
Benzoxazinas/administração & dosagem , Agonistas de Receptores de Canabinoides/administração & dosagem , Canabinoides/administração & dosagem , Morfolinas/administração & dosagem , Naftalenos/administração & dosagem , Neuropeptídeos/administração & dosagem , Medição da Dor/efeitos dos fármacos , Animais , Sinergismo Farmacológico , Quimioterapia Combinada , Masculino , Camundongos , Medição da Dor/métodos
10.
PLoS One ; 9(8): e103773, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25090615

RESUMO

Neuropeptide FF (NPFF) is known to be an endogenous opioid-modulating peptide. Nevertheless, very few researches focused on the interaction between NPFF and endogenous opioid peptides. In the present study, we have investigated the effects of NPFF system on the supraspinal antinociceptive effects induced by the endogenous µ-opioid receptor agonists, endomorphin-1 (EM-1) and endomorphin-2 (EM-2). In the mouse tail-flick assay, intracerebroventricular injection of EM-1 induced antinociception via µ-opioid receptor while the antinociception of intracerebroventricular injected EM-2 was mediated by both µ- and κ-opioid receptors. In addition, central administration of NPFF significantly reduced EM-1-induced central antinociception, but enhanced EM-2-induced central antinociception. The results using the selective NPFF1 and NPFF2 receptor agonists indicated that the EM-1-modulating action of NPFF was mainly mediated by NPFF2 receptor, while NPFF potentiated EM-2-induecd antinociception via both NPFF1 and NPFF2 receptors. To further investigate the roles of µ- and κ-opioid systems in the opposite effects of NPFF on central antinociception of endomprphins, the µ- and κ-opioid receptors selective agonists DAMGO and U69593, respectively, were used. Our results showed that NPFF could reduce the central antinociception of DAMGO via NPFF2 receptor and enhance the central antinociception of U69593 via both NPFF1 and NPFF2 receptors. Taken together, our data demonstrate that NPFF exerts opposite effects on central antinociception of endomorphins and provide the first evidence that NPFF potentiate antinociception of EM-2, which might result from the interaction between NPFF and κ-opioid systems.


Assuntos
Nociceptividade/efeitos dos fármacos , Oligopeptídeos/farmacologia , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Benzenoacetamidas/farmacologia , Dipeptídeos/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Injeções Intraventriculares , Masculino , Camundongos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Pirrolidinas/farmacologia
11.
Neurosci Lett ; 578: 187-91, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25010019

RESUMO

The nonapeptide hemopressin and its N-terminal extension VD-hemopressin(α) were reported as an antagonist/inverse agonist and an agonist of CB1 receptor, respectively. These novel cannabinoid peptides have been demonstrated to modulate the acute pain. In the present study, hemopressin (11, 22 and 45 nmol, i.c.v.) dose-dependently produced antinociception after supraspinal administration in the radiant heat tail-flick test. Furthermore, the development of antinociceptive tolerance to hemopressin, VD-hemopressin(α) and WIN55,212-2, and cross-tolerance among these cannabinoids were investigated in mice. The tolerance developed on day 4 after supraspinal injection of hemopressin (45 nmol), VD-hemopressin(α) (20 nmol) and WIN55,212-2 (7.5 nmol). Our results indicated symmetrical cross-tolerance between hemopressin, VD-hemopressin(α) and WIN55,212-2 at the supraspinal level in mice. These results demonstrate that both hemopressin and VD-hemopressin(α) have a time-course and extent of tolerance similar to the synthetic cannabinoid WIN55,212-2. In addition, our data imply that a common mechanism is involved in the antinociception of the three cannabinoid ligands.


Assuntos
Analgésicos/farmacologia , Benzoxazinas/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Hemoglobinas/farmacologia , Morfolinas/farmacologia , Naftalenos/farmacologia , Nociceptividade/efeitos dos fármacos , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Agonismo Inverso de Drogas , Tolerância a Medicamentos , Ligantes , Masculino , Camundongos , Medula Espinal , Cauda
12.
Peptides ; 56: 45-51, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24681436

RESUMO

Previous studies suggest that cannabinoids system plays an important role in cardiovascular regulation. (m)VD-hemopressin(α) (VD-Hpα), an 11-residue peptide originating from the α1 chain of hemoglobin, was recently reported as a selective agonist of cannabinoid CB1 receptor. The present study was undertaken to investigate the intrathecal (i.t.) action of (m)VD-Hpα on blood pressure in urethane-anesthetized rats. Our results demonstrated that injections of (m)VD-Hpα (5-30 nmol, i.t.) produced a dose-dependent decrease in mean arterial pressure (MAP), similar to that of the non-peptidic cannabinoid receptor agonist WIN55212-2 (1.25-10 nmol, i.t.). The hypotensive effect of (m)VD-Hpα was not influenced by the CB1 receptor antagonist AM251 (20 nmol, i.t.) or the CB2 receptor antagonist AM630 (20 nmol, i.t.). However, WIN55212-2-induced hypotension was almost completely prevented by i.t. administration of AM251, not by AM630. The spinal hypotension of (m)VD-Hpα and WIN55212-2 was significantly reduced by pretreatment with the α-adrenoceptor antagonist phentolamine (1 mg/kg, i.v.), but not by the ß-adrenoceptor antagonist propranolol (2 mg/kg, i.v.) or the muscarinic receptor antagonist atropine (2 mg/kg, i.v.). In addition, L-NAME (50 mg/kg, i.v.), the inhibitor of nitric oxide (NO) synthase, significantly reduced WIN55212-2-induced hypotension, but had no effect on the hypotensive response to (m)VD-Hpα. Collectively, the results show that i.t. administration of (m)VD-Hpα induces a decrease in MAP via a non-CB1 and non-CB2 mechanism.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hemoglobinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Uretana/farmacologia , Animais , Hemoglobinas/administração & dosagem , Humanos , Hipotensão/induzido quimicamente , Injeções Espinhais , Masculino , Óxido Nítrico/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Wistar , Receptor CB2 de Canabinoide/metabolismo
13.
J Pharmacol Exp Ther ; 348(2): 316-23, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307201

RESUMO

The cannabinoid system has been demonstrated to modulate the acute and chronic pain of multiple origins. Mouse VD-hemopressin(α) [(m)VD-Hpα], an 11-residue α-hemoglobin-derived peptide, was recently reported to function as a selective agonist of the cannabinoid receptor type 1 (CB1) in vitro. To characterize its behavioral and physiological properties, we investigated the in vivo effects of (m)VD-Hpα in mice. In the mouse tail-flick test, (m)VD-Hpα dose-dependently induced antinociception after supraspinal (EC50 = 6.69 nmol) and spinal (EC50 = 2.88 nmol) administration. The antinociceptive effects of (m)VD-Hpα (intracerebroventricularly and intrathecally) were completely blocked by N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3- carboxamide (AM251; CB1 antagonist), but not by 6-iodo-2-methyl-1-[2-(4-morpholinyl)ethyl]-1H-indol-3-yl(4-methoxyphenyl)-methanone (AM630; CB2 antagonist) or naloxone (opioid antagonist), showing its selectivity to the CB1 receptor. Furthermore, the central nervous system (CNS) effects of (m)VD-Hpα were evaluated in body temperature, locomotor activity, tolerance development, reward, and food intake assays. At the highly antinociceptive dose (3 × EC50), (m)VD-Hpα markedly exerted hypothermia and hypoactivity after supraspinal administration. Repeated intracerebroventricular injection of (m)VD-Hpα resulted in both development of tolerance to antinociception and conditioned place aversion. In addition, central injection of (m)VD-Hpα dose-dependently stimulated food consumption. These findings demonstrate that this novel cannabinoid peptide agonist induces CB1-mediated central antinociception with some CNS effects, which further supports a CB1 agonist character of (m)VD-Hpα. Moreover, the current study will be helpful to understand the in vivo properties of the endogenous peptide agonist of the cannabinoid CB1 receptor.


Assuntos
Analgésicos/administração & dosagem , Agonistas de Receptores de Canabinoides/administração & dosagem , Sistema Nervoso Central/efeitos dos fármacos , Hemoglobinas/administração & dosagem , Proteínas do Tecido Nervoso/agonistas , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Receptor CB1 de Canabinoide/agonistas , Analgésicos/efeitos adversos , Analgésicos/antagonistas & inibidores , Analgésicos/uso terapêutico , Animais , Regulação do Apetite/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Regulação da Temperatura Corporal/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/efeitos adversos , Agonistas de Receptores de Canabinoides/química , Agonistas de Receptores de Canabinoides/uso terapêutico , Antagonistas de Receptores de Canabinoides/efeitos adversos , Antagonistas de Receptores de Canabinoides/farmacologia , Sistema Nervoso Central/metabolismo , Hemoglobinas/efeitos adversos , Hemoglobinas/química , Hemoglobinas/uso terapêutico , Infusões Intraventriculares , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos , Antagonistas de Entorpecentes/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Oligopeptídeos/uso terapêutico , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/metabolismo , Receptores Opioides/metabolismo
14.
Behav Brain Res ; 248: 51-6, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23578757

RESUMO

It has been demonstrated that the endogenous mu opioid (MOP) agonist endomorphin-2 (EM-2) produces conditioned place aversion (CPA) and in contrast, morphine exerts opposite action. Neuropeptide FF (NPFF) was reported to act as a functional antagonist of mu opioid receptor and to exert opioid-modulating activities. The present study examined the influence of NPFF on the rewarding action of EM-2, using the unbiased conditioned place preference (CPP) paradigm. For testing the effect of NPFF on the acquisition of EM-2-induced CPA, NPFF and EM-2 were co-injected on the conditioning days without drug treatment on the followed test day. To explore the effect of NPFF on the expression of EM-2-induced CPA, EM-2 was administered alone on the conditioning days, and NPFF was given 5 min before placement in the CPP apparatus on the test day. The results showed that NPFF (2.5, 5 and 10 nmol, i.c.v.) alone caused little place preference change. However, NPFF dose-dependently reversed the acquisition of CPA induced by 30 nmol EM-2 (i.c.v.). Similarly, the expression of EM-2-induced CPA was also reduced by NPFF. Moreover, the effects of NPFF on the acquisition and the expression of EM-2-induced CPA were completely blocked by the NPFF receptors antagonist RF9 (10 nmol, i.c.v.). However, central injection of NPFF neither changed the locomotor activity nor modified the locomotor action of EM-2. These data provide the first evidence for a functional interaction of the endogenous ligands for NPFF and MOP receptors, and further support an anti-opioid character of NPFF system.


Assuntos
Oligopeptídeos/farmacologia , Receptores de Neuropeptídeos/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Animais , Condicionamento Psicológico/fisiologia , Camundongos , Morfina/administração & dosagem , Morfina/farmacologia , Oligopeptídeos/administração & dosagem , Peptídeos Opioides/farmacologia , Recompensa
15.
Behav Brain Res ; 233(2): 428-33, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22659392

RESUMO

Neuropeptide FF (NPFF) belongs to a neuropeptide family including two receptors (NPFF(1) and NPFF(2)). NPFF system has been reported to play important roles in pain transmission. The aim of the present study was to investigate the roles of NPFF related peptides and their receptors in swim stress-induced analgesia (SIA). Nociceptive test was performed in mice stressed by forced swimming in water at 15 °C (cold water swimming) or 32 °C (warm water swimming). Warm water swimming produced a naloxone-mediated antinociceptive effect. This warm water swim SIA was dose-dependently antagonized by i.c.v. injection of NPFF and two related peptides (3-30 nmol), NPVF and dNPA, which exhibited the highest selectivities for NPFF(1) and NPFF(2) receptors, respectively. Moreover, the selective NPFF receptor antagonist RF9 (30 nmol) was inactive by itself, but prevented the effects of NPFF and related peptides. Cold-water swimming produced a wilder analgesic effect that was blocked by MK-801, but not naloxone. However, NPFF system failed to modify the cold water swim stress-induced analgesia. These findings demonstrated that NPFF and related peptides attenuated opioid-mediated form of SIA via NPFF receptors in the brain, but not non-opioid swim stress-induced analgesia. These data further support an anti-opioid character of NPFF system.


Assuntos
Analgesia , Temperatura Baixa , Temperatura Alta , Oligopeptídeos/administração & dosagem , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/fisiopatologia , Adamantano/administração & dosagem , Adamantano/análogos & derivados , Análise de Variância , Animais , Dipeptídeos/administração & dosagem , Maleato de Dizocilpina/administração & dosagem , Relação Dose-Resposta a Droga , Injeções Intra-Articulares , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Neuropeptídeos/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Estresse Psicológico/etiologia , Natação/psicologia
16.
Neuropharmacology ; 62(2): 855-64, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21945715

RESUMO

It has been demonstrated that opioid and cannabinoid receptor systems can produce similar signal transduction and behavioural effects. Neuropeptide FF (NPFF) belongs to an opioid-modulating peptide family. NPFF has been reported to play important roles in control of pain and analgesia through interactions with the opioid system. We were interested in whether the central and peripheral antinociception of cannabinoids could be influenced by supraspinal NPFF system. The present study examined the effects of NPFF and related peptides on the antinociceptive activities induced by the non-selective cannabinoid receptors agonist WIN55,212-2, given by supraspinal and intraplantar routes. In mice, the central and peripheral antinociception of WIN55,212-2 are mediated by cannabinoid CB(1) and CB(2) receptors, respectively. Interestingly, central administration of NPFF significantly reduced central and peripheral analgesia of cannabinoids in dose-dependent manners. In contrast, dNPA and NPVF (i.c.v.), two highly selective agonists for NPFF(2) and NPFF(1) receptors, dose-dependently augmented the antinociception caused by intracerebroventricular and intraplantar injection of WIN55,212-2. Additionally, pretreatment with the NPFF receptors selective antagonist RF9 (i.c.v.) markedly reduced the cannabinoid-modulating activities of NPFF and related peptides in nociceptive assays. These data provide the first evidence for a functional interaction between NPFF and cannabinoid systems, indicating that activation of central NPFF receptors interferes with cannabinoid-mediated central and peripheral antinociception. Intriguingly, the present work may pave the way for a new strategy of using combination treatment of cannabinoid and NPFF agonists for pain management. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Assuntos
Oligopeptídeos/metabolismo , Medição da Dor/efeitos dos fármacos , Dor/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Analgesia , Analgésicos/farmacologia , Animais , Benzoxazinas/farmacologia , Indóis/farmacologia , Masculino , Camundongos , Morfolinas/farmacologia , Naloxona/farmacologia , Naftalenos/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia
17.
Protein Pept Lett ; 18(4): 403-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21121889

RESUMO

Neuropeptide FF (NPFF) belongs to an opioid-modulating peptide family. NPFF has been reported to play important roles in the control of pain and analgesia through interactions with the opioid system. However, very few studies examined the effect of supraspinal NPFF system on analgesia induced by opiates administered at the peripheral level. In the present study, intracerebroventricular (i.c.v.) injection of NPFF (1, 3 and 10 nmol) dose-dependently inhibited systemic morphine (0.12 mg, i.p.) analgesia in the mouse tail flick test. Similarly, i.c.v. administration of dNPA and NPVF, two agonists highly selective for NPFF(2) and NPFF(1) receptors, respectively, decreased analgesia induced by i.p. morphine in mice. Furthermore, these anti-opioid activities of NPFF and related peptides were blocked by pretreatment with the NPFF receptors selective antagonist RF9 (10 nmol, i.c.v.). These results demonstrate that activation of central NPFF(1) and NPFF(2) receptors has the similar anti-opioid actions on the antinociceptive effect of systemic morphine.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Morfina/antagonistas & inibidores , Antagonistas de Entorpecentes/administração & dosagem , Oligopeptídeos/administração & dosagem , Analgesia , Animais , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Masculino , Camundongos , Camundongos Endogâmicos , Morfina/administração & dosagem , Neuropeptídeos/administração & dosagem , Dor/tratamento farmacológico , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA