Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci Methods ; 386: 109783, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36610617

RESUMO

Pharmacological assays based on the measurement of nociceptive responses in laboratory animals are a fundamental tool to assess analgesic strategies. During our experience with this type of experiments, we have been repeatedly challenged by different concerns related to their interpretation or relevance. Although these subjects are frequently discussed in our lab, they do not usually find a place in research articles with original data, in which the focus on results seems mandatory. In the present manuscript we try to discuss as central issues some of these aspects that often cross transversally our research. We have gathered them in five topics inspired by the results obtained in our laboratory. The two initial sections are devoted to the influence of the behavioral method used to assess nociception on the results achieved, as well as to the possibility that data may be more easily accepted when obtained with standard methods than with alternative ones. The third topic is related to the difficulties encountered when working with a molecule that may evoke dual effects, acting as pronociceptive or antinociceptive depending on the dose. The fourth point deals with the situation in which a particular hyperalgesic reaction is related to several molecules but the single inhibition of only one of them can completely prevent it. Finally, the last issue is addressed to comment the impact in the progress of pain research of experiments performed in animal models of pathological settings.


Assuntos
Hiperalgesia , Dor , Animais , Medição da Dor , Analgésicos/farmacologia
2.
Sci Rep ; 10(1): 18599, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33122720

RESUMO

The newly designed fentanyl derivative [( ±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide] (NFEPP) was recently shown to produce analgesia selectively via peripheral mu-opioid receptors (MOR) at acidic pH in rat inflamed tissues. Here, we examined the pH-dependency of NFEPP binding to brain MOR and its effects on bone cancer-induced pain in mice. The IC50 of NFEPP to displace bound [3H]-DAMGO was significantly higher compared to fentanyl at pH 7.4, but no differences were observed at pH 5.5 or 6.5. Intravenous NFEPP (30-100 nmol/kg) or fentanyl (17-30 nmol/kg) inhibited heat hyperalgesia in mice inoculated with B16-F10 melanoma cells. The peripherally-restricted opioid receptor antagonist naloxone-methiodide reversed the effect of NFEPP (100 nmol/kg), but not of fentanyl (30 nmol/kg). The antihyperalgesic effect of NFEPP was abolished by a selective MOR- (cyprodime), but not delta- (naltrindole) or kappa- (nor-binaltorphimine) receptor antagonists. Ten-fold higher doses of NFEPP than fentanyl induced maximal antinociception in mice without tumors, which was reversed by the non-restricted antagonist naloxone, but not by naloxone-methiodide. NFEPP also reduced heat hyperalgesia produced by fibrosarcoma- (NCTC 2472) or prostate cancer-derived (RM1) cells. These data demonstrate the increased affinity of NFEPP for murine MOR at low pH, and its ability to inhibit bone cancer-induced hyperalgesia through peripheral MOR. In mice, central opioid receptors may be activated by ten-fold higher doses of NFEPP.


Assuntos
Analgésicos Opioides/farmacologia , Dor do Câncer/tratamento farmacológico , Piperidinas/farmacologia , Receptores Opioides mu/metabolismo , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/metabolismo , Dor do Câncer/metabolismo , Linhagem Celular Tumoral , Fentanila/farmacologia , Concentração de Íons de Hidrogênio , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Ligantes , Masculino , Melanoma Experimental/complicações , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Morfinanos/farmacologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia
3.
Cell Mol Neurobiol ; 39(8): 1115-1124, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31203533

RESUMO

Apart from its involvement in immune functions, the chemokine CCL1 can participate in the modulation of nociceptive processing. Previous studies have demonstrated the hypernociceptive effect produced by CCL1 in the spinal cord, but its possible action on peripheral nociception has not yet been characterized. We describe here that the subcutaneous administration of CCL1 (1-10 µg/kg) produces dose-dependent and long-lasting increases in thermal withdrawal latencies measured by the unilateral hot plate test in mice. The antinociceptive nature of this effect is further supported by the reduction of spinal neurons expressing Fos protein in response to a noxious thermal stimulus observed after the administration of 10 µg/kg of CCL1. CCL1-induced antinociception was inhibited after systemic, but not spinal administration of the selective antagonist R243 (0.1-1 mg/kg), demonstrating the participation of peripheral CCR8 receptors. The absence of this analgesic effect in mice treated with a dose of cyclophosphamide that produces a drastic depletion of leukocytes suggests its dependency on white blood cells. Furthermore, whereas the antinociceptive effect of CCL1 was unaffected after the treatment with either the antagonist of opioid receptors naloxone or the cannabinoid type 1 receptor blocker AM251, it was dose-dependently inhibited after the administration of the CB2 receptor antagonist SR144528 (0.1-1 mg/kg). The detection by ELISA of an increased presence of the endocannabinoid 2-arachidonoylglycerol after the administration of an analgesic dose of CCL1 supports the notion that CCL1 can evoke thermal analgesia through the release of this endocannabinoid from circulating leukocytes.


Assuntos
Analgesia , Quimiocina CCL1/administração & dosagem , Endocanabinoides/metabolismo , Temperatura , Analgésicos/farmacologia , Animais , Ácidos Araquidônicos/metabolismo , Ciclofosfamida , Glicerídeos/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/metabolismo , Masculino , Camundongos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Receptores CCR8/metabolismo
4.
Scand J Pain ; 14: 25-38, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28850427

RESUMO

BACKGROUND: The first line pharmacological treatment of cancer pain is morphine and surrogates but a significant pain relief and a reduction of the side-effects of these compounds makes it necessary to combine them with other drugs acting on different targets. The aim of this study was to measure the antinociceptive effect on cancer-induced bone pain resulting from the association of the endogenous opioids enkephalin and non-opioid analgesic drugs. For this purpose, PL265 a new orally active single dual inhibitor of the two degrading enkephalins enzymes, neprilysin (NEP) and aminopeptidase N (APN) was used. It strictly increased the levels of enkephalin at their sites of releases. The selected non-opioid compounds are: gabapentin, A-317491 (P2X3 receptor antagonist), ACEA (CB1 receptor antagonist), AM1241 (CB2 receptor antagonist), JWH-133 (CB2 receptor antagonist), URB937 (FAAH inhibitor), and NAV26 (Nav1.7 channel blocker). METHODS: Experiments. Experiments were performed in 5-6 weeks old (26-33g weight) C57BL/6 mice. Cell culture and cell inoculation. B16-F10 melanoma cells were cultured and when preconfluent, treated and detached. Finally related cells were resuspended to obtain a concentration of 2×106 cells/100µL. Then 105 cells were injected into the right tibial medullar cavity. Control mice were treated by killed cells by freezing. Behavioural studies. Thermal withdrawal latencies were measured on a unilatered hot plate (UHP) maintained at 49±0.2°C. Mechanical threshold values were obtained by performing the von Frey test using the "up and down" method. To evaluate the nature (additive or synergistic) of the interactions between PL265 and different drugs, an isobolographic analysis following the method described by Tallarida was performed. RESULTS: The results demonstrate the ability of PL265, a DENKI that prevents the degradation of endogenous ENKs, to counteract cancer-induced bone thermal hyperalgesia in mice, by exclusively stimulating peripheral opioid receptors as demonstrated by used of an opioid antagonist unable to enter the brain. The development of such DENKIs, endowed with druggable pharmacokinetic characteristics, such as good absorption by oral route, can be considered as an important step in the development of much needed novel antihyperalgesic drugs. Furthermore, all the tested combinations resulted in synergistic antihyperalgesic effects. As shown here, the greatest synergistic antinociceptive effect (doses could be lowered by 70%) was produced by the combination of PL265 with the P2X3 receptor antagonist (A-317491), cannabinoid CB1 receptor agonist (exogenous, ACEA and endogenous URB937-protected-AEA) and Nav1.7 blocker (NAV26) whose mechanism of action involves the direct activation of the enkephalinergic system. CONCLUSIONS: These multi-target-based antinociceptive strategies using combinations of non-opioid drugs with dual inhibitors of enkephalin degrading enzymes may bring therapeutic advantages in terms of efficacy and safety by allowing the reduction of doses of one of the compounds or of both, which is of the utmost interest in the chronic treatment of cancer pain. IMPLICATIONS: This article presents synergistic antinociceptive effect produced by the combination of PL265 with non-opioid analgesic drugs acting via unrelated mechanisms. These multi-target-based antinociceptive strategies may bring therapeutic advantages by allowing the reduction of doses, which is of great interest in the chronic treatment of cancer pain.


Assuntos
Analgésicos/farmacologia , Osso e Ossos/efeitos dos fármacos , Dor do Câncer/tratamento farmacológico , Neprilisina/antagonistas & inibidores , Propionatos/farmacologia , Administração Oral , Animais , Osso e Ossos/fisiopatologia , Dor do Câncer/fisiopatologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Encefalinas/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Transplante de Neoplasias , Neprilisina/metabolismo , Distribuição Aleatória
6.
Prostate ; 75(1): 70-83, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25263196

RESUMO

BACKGROUND: Pain due to bone metastases of prostatic origin is a relevant clinical issue. We study here the nociceptive responses obtained in mice receiving the intratibial inoculation of RM1 prostate cancer cells. METHODS: 10(2) -10(5) RM1 cells were inoculated to C57BL/6 mice and tumor development was analysed histologically and with luciferase-expressing RM1 cells. Spinal astroglial (GFAP) or microglial (Iba-1) expression was assessed with immunohistochemical methods and hypernociception was measured by the unilateral hot plate, the paw pressure and the von Frey tests. The analgesic effect of morphine, zoledronic acid or the CCR2 antagonist RS504393 was measured. Levels of the chemokines CCL2, CCL3, and CCL5 were determined by ELISA. RESULTS: The inoculation of 10(3) RM1 cells induced tumoral growth in bone with a mixed osteoclastic/osteoblastic pattern and evoked astroglial, but not microglial, activation in the spinal cord. Hyperalgesia and allodynia were already established four days after inoculation and dose-dependently inhibited by the s.c. administration of morphine (1-5 mg/kg) or zoledronic acid (1-3 mg/kg). CCL2 and CCL5, but not CCL3, were released by RM1 cells in culture whereas only an increased presence of CCL2 was found in bone tumor homogenates. The administration of the CCR2 antagonist RS504393 (0.3-3 mg/kg) inhibited RM1 induced thermal hyperalgesia without modifying mechanical allodynia. CONCLUSION: The intratibial inoculation of RM1 cells in immunocompetent mice induces hypernociceptive responses and can be useful to perform studies of bone cancer induced pain related to androgen-independent prostate cancer. The antinociceptive role derived from the blockade of the CCR2 chemokine receptors is further envisaged.


Assuntos
Neoplasias Ósseas/secundário , Hiperalgesia/fisiopatologia , Dor Nociceptiva/fisiopatologia , Neoplasias da Próstata/patologia , Tíbia/patologia , Animais , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Difosfonatos/farmacologia , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Proteína Glial Fibrilar Ácida , Hiperalgesia/tratamento farmacológico , Imidazóis/farmacologia , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Morfina/farmacologia , Transplante de Neoplasias , Proteínas do Tecido Nervoso , Dor Nociceptiva/tratamento farmacológico , Receptores CCR2/metabolismo , Medula Espinal/patologia , Ácido Zoledrônico
7.
Cell Mol Neurobiol ; 34(1): 143-56, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24122510

RESUMO

The hypernociceptive role played by the chemokine CCL2, and its main receptor, CCR2, in pathological settings is being increasingly recognized. We aimed to characterize the involvement of spinal CCL2 in the hyperalgesia due to the intratibial inoculation of fibrosarcoma NCTC 2472 cells in mice. The intrathecal (i.t.) administration of the CCR2 antagonist RS 504393 (1­3 µg) or an anti-CCL2 antibody inhibited tumoral hyperalgesia. No change in the expression of spinal CCR2 was detected by western blot, whereas immunohistochemical experiments demonstrated increased CCL2 staining at the superficial laminae of the spinal cord ipsilateral to the tumor. This spinal CCL2 does not seem to be released from nociceptors since CCL2 mRNA and CCL2 levels in DRGs, as measured by RT-PCR and ELISA, remain unmodified in tumor-bearing mice. In contrast, immunohistochemical assays demonstrated the spinal up-regulations of GFAP and Iba-1, respective markers of astroglia and microglia, and the expression of CCL2 in both types of glial cells at the superficial laminae of the spinal cord of tumor-bearing mice. Finally, since CCL2 could induce astroglial or microglial activation, we studied whether the blockade of CCR2 could inhibit the increased spinal glial expression. GFAP, but not Iba-1, up-regulation was reduced in tumor-bearing mice treated for 3 days with i.t. RS 504393, indicating that spinal CCL2 acts as an astroglial activator in this setting. The participation at spinal level of CCL2/CCR2 in tumoral hypernociception, together with its previously described involvement at periphery, makes attractive the modulation of this system for the alleviation of neoplastic pain.


Assuntos
Astrócitos/metabolismo , Neoplasias Ósseas/complicações , Quimiocina CCL2/metabolismo , Hiperalgesia/etiologia , Microglia/metabolismo , Osteossarcoma/complicações , Medula Espinal/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Benzoxazinas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Ensaio de Imunoadsorção Enzimática , Gânglios Espinais/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Camundongos , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Compostos de Espiro/farmacologia , Regulação para Cima/efeitos dos fármacos
8.
Brain Res Bull ; 95: 21-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23562605

RESUMO

The antineoplastic paclitaxel induces a sensory neuropathy that involves the spinal release of neuroinflammatory mediators and activation of glial cells. Although the chemokine CCL2 can evoke glial activation and its participation in neuropathic pain has been demonstrated in other models, its involvement in paclitaxel-evoked neuropathy has not been previously explored. Paclitaxel-evoked cold hypernociception was assessed in mice by the unilateral cold plate test and the effects on cold hyperalgesia of the CCR2 antagonist RS 504393, the CCR1 antagonist J113863, the microglial inhibitor minocycline or an anti-CCL2 antibody were tested. Furthermore, ELISA measurements of CCL2 concentration and immunohistochemical assays of Iba-1 and GFAP, markers of microglial and astroglial cells respectively, were performed in the lumbar spinal cord. Cold hypernociception measured 3 days after the administration of paclitaxel (10mg/kg) was inhibited by the s.c. (0.3-3mg/kg) or i.t. (1-10 µg) administration of RS 504393 but not of J113863 (3-30 mg/kg). CCL2 levels measured by ELISA in the lumbar spinal cord were augmented in mice treated with paclitaxel and the i.t. administration of an anti-CCL2 antibody completely suppressed paclitaxel-evoked cold hyperalgesia, strongly suggesting that CCL2 is involved in the hypernociception evoked by this taxane. Besides, the implication of microglial activation is supported by the increase in the immunolabelling of Iba-1, but not GFAP, in the spinal cord of paclitaxel-treated mice and by the inhibition of cold hyperalgesia produced by the i.t. administration of the microglial inhibitor minocycline (1-10 nmol). Finally, the neutralization of spinal CCL2 by the i.t. administration of a selective antibody for 3 days almost totally inhibited paclitaxel-evoked microglial activation. In conclusion, our results indicate that paclitaxel-evoked cold hypernociception depends on the activation of CCR2 due to the spinal release of CCL2 and the subsequent microglial activation.


Assuntos
Quimiocina CCL2/biossíntese , Hiperalgesia/induzido quimicamente , Microglia/metabolismo , Neuralgia/metabolismo , Paclitaxel/farmacologia , Medula Espinal/metabolismo , Animais , Temperatura Baixa , Hiperalgesia/metabolismo , Masculino , Camundongos , Minociclina/farmacologia , Neuralgia/induzido quimicamente , Receptores CCR2/metabolismo , Medula Espinal/citologia
9.
Naunyn Schmiedebergs Arch Pharmacol ; 385(11): 1053-61, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22976830

RESUMO

The participation of the chemokine CCL2 (monocyte chemoattractant protein-1) in inflammatory and neuropathic pain is well established. Furthermore, the release of CCL2 from a NCTC 2472 cells-evoked tumor and its involvement in the upregulation of calcium channel α2δ1 subunit of nociceptors was demonstrated. In the present experiments, we have tried to determine whether the increase in CCL2 levels is a common property of painful tumors and, in consequence, the administration of a chemokine receptor type 2 (CCR2) antagonist can inhibit tumoral hypernociception. CCL2 levels were measured by ELISA in the tumoral region of mice intratibially inoculated with NCTC 2472 or B16-F10 cells, and the antihyperalgesic and antiallodynic effects evoked by the administration of the selective CCR2 antagonist RS 504393 were assessed. Cultured NCTC 2472 cells release CCL2 and their intratibial inoculation evokes the development of a tumor in which CCL2 levels are increased. Moreover, the systemic or peritumoral administration of RS 504393 inhibited thermal and mechanical hyperalgesia, but not mechanical allodynia evoked after the inoculation of these cells. Thermal hyperalgesia was also inhibited by the peritumoral administration of a neutralizing CCL2 antibody. In contrast, no change in CCL2 levels was observed in mice inoculated with B16-F10 cells, and RS 504393 did not inhibit the hypernociceptive reactions evoked by their intratibial inoculation. The peripheral release of CCL2 is involved in the development of thermal and mechanical hyperalgesia, but not mechanical allodynia evoked by the inoculation of NCTC 2472 cells, whereas this chemokine seems unrelated to the hypernociception induced by B16-F10 cells.


Assuntos
Benzoxazinas/farmacologia , Neoplasias Ósseas/complicações , Quimiocina CCL2/metabolismo , Hiperalgesia/patologia , Receptores CCR2/antagonistas & inibidores , Compostos de Espiro/farmacologia , Animais , Benzoxazinas/administração & dosagem , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fibrossarcoma/patologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Compostos de Espiro/administração & dosagem , Tíbia
10.
J Pharmacol Exp Ther ; 341(3): 634-45, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22393248

RESUMO

Small peptides patterned after the N terminus of the synaptosomal protein of 25 kDa, a member of the protein complex implicated in Ca(2+)-dependent neuronal exocytosis, inhibit in vitro the release of neuromodulators involved in pain signaling, suggesting an in vivo analgesic activity. Here, we report that compound DD04107 (palmitoyl-EEMQRR-NH(2)), a 6-mer palmitoylated peptide that blocks the inflammatory recruitment of ion channels to the plasma membrane of nociceptors and the release of calcitonin gene-related peptide from primary sensory neurons, displays potent and long-lasting in vivo antihyperalgesia and antiallodynia in chronic models of inflammatory and neuropathic pain, such as the complete Freund's adjuvant, osteosarcoma, chemotherapy, and diabetic neuropathic models. Subcutaneous administration of the peptide produced a dose-dependent antihyperalgesic and antiallodynic activity that lasted ≥24 h. The compound showed a systemic distribution, characterized by a bicompartmental pharmacokinetic profile. Safety pharmacology studies indicated that the peptide is largely devoid of side effects and substantiated that the in vivo activity is not caused by locomotor impairment. Therefore, DD04107 is a potent and long-lasting antinociceptive compound that displays a safe pharmacological profile. These findings support the notion that neuronal exocytosis of receptors and neuronal algogens pivotally contribute to chronic inflammatory and neuropathic pain and imply a central role of peptidergic nociceptor sensitization to the pathogenesis of pain.


Assuntos
Analgésicos/farmacologia , Modelos Animais de Doenças , Exocitose/efeitos dos fármacos , Inflamação/tratamento farmacológico , Lipopeptídeos/farmacologia , Neuralgia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Analgésicos/efeitos adversos , Analgésicos/farmacocinética , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Carragenina/toxicidade , Relação Dose-Resposta a Droga , Hiperalgesia/tratamento farmacológico , Injeções Subcutâneas , Lipopeptídeos/efeitos adversos , Lipopeptídeos/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Experimentais/patologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Fatores de Tempo
11.
Fundam Clin Pharmacol ; 26(3): 363-72, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21306432

RESUMO

Agonists of µ-opioid receptors are currently used in the management of cancer pain. However, several data suggest that the analgesic effect of morphine can diminish during the development of experimental tumors. By using a thermal test, we have studied whether the analgesic effect evoked by morphine is altered in mice bearing two painful bone tumors. The analgesic effect evoked by systemic morphine remained unaltered after the intratibial inoculation of B16-F10 melanoma cells and was potentiated after the inoculation of NCTC 2472 osteosarcoma cells. Although the number of spinal µ-opioid receptors measured by western blot studies was not augmented in osteosarcoma-bearing mice, the analgesia evoked by intrathecal (i.t.) morphine was also enhanced. The analgesic response produced by the spinal administration of the Gi/o protein activator mastoparan was amplified, whereas the analgesic response evoked by the i.t. administration of the N-type calcium channel blocker ω-conotoxin remained unaltered. The efficacy of the GIRK channel blocker tertiapin-Q to antagonize the analgesic effect produced by a maximal dose of morphine was also increased in osteosarcoma-bearing mice. Our results seem to indicate that the analgesic effect of morphine on thermal nociception can be enhanced in response to the development of particular bone tumors in mice, being this potentiation probably related to a greater efficacy of the transduction system driven by Gi/o proteins and GIRK channels.


Assuntos
Analgesia/métodos , Neoplasias Ósseas/tratamento farmacológico , Temperatura Alta , Morfina/administração & dosagem , Medição da Dor/métodos , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Temperatura Alta/efeitos adversos , Masculino , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Dor/tratamento farmacológico , Dor/metabolismo , Dor/patologia , Receptores Opioides mu/biossíntese
12.
Eur J Pharmacol ; 668(1-2): 184-9, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21771590

RESUMO

The stimulation of spinal cannabinoid type 2 (CB(2)) receptors is a suitable strategy for the alleviation of experimental pain symptoms. Several reports have described the up-regulation of spinal cannabinoid CB(2) receptors in neuropathic settings together with the analgesic effects derived from their activation. Besides, we have recently reported in two murine bone cancer models that the intrathecal administration of cannabinoid CB(2) receptor agonists completely abolishes hyperalgesia and allodynia, whereas spinal cannabinoid CB(2) receptor expression remains unaltered. The present experiments were designed to measure the expression of spinal cannabinoid CB(2) receptors as well as the analgesic efficacy derived from their stimulation in mice chronically inflamed by the intraplantar injection of complete Freund's adjuvant 1 week before. Both spinal cannabinoid CB(2) receptors mRNA measured by real-time PCR and cannabinoid CB(2) receptor protein levels measured by western blot remained unaltered in inflamed mice. Besides, the intrathecal (i.t.) administration of the cannabinoid CB(2) receptor agonists AM1241, (R,S)-3-(2-Iodo-5-nitrobenzoyl)-1-(1-methyl-2-piperidinylmethyl)-1H-indole, (0.03-1 µg) and JWH 133, (6aR,10aR)-3-(1,1-Dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran, (3-30 µg) dose-dependently blocked inflammatory thermal hyperalgesia and mechanical allodynia. The analgesic effects induced by both agonists were counteracted by the coadministration of the selective cannabinoid CB(2) receptor antagonist SR144528, 5-(4-chloro-3-methylphenyl)-1-[(4-methylphenyl)methyl]-N-[(1S,2S,4R)-1,3,3-trimethylbicyclo[2.2.1]hept-2-yl]-1H-pyrazole-3-carboxamide, (5 µg) but not by the cannabinoid CB(1) receptor antagonist AM251, N-(Piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide, (10 µg). The effects induced by AM1241 were also inhibited by the coadministration of the opioid receptor antagonist, naloxone (1 µg). These results demonstrate that effective analgesia can be achieved in chronic inflammatory settings through the stimulation of spinal cannabinoid CB(2) receptors even if this receptor population is not up-regulated.


Assuntos
Analgésicos/farmacologia , Inflamação/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Analgésicos/administração & dosagem , Analgésicos/uso terapêutico , Animais , Canabinoides/administração & dosagem , Canabinoides/farmacologia , Canabinoides/uso terapêutico , Doença Crônica/tratamento farmacológico , Adjuvante de Freund/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Inflamação/induzido quimicamente , Inflamação/complicações , Inflamação/tratamento farmacológico , Masculino , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor CB2 de Canabinoide/agonistas , Temperatura
13.
Naunyn Schmiedebergs Arch Pharmacol ; 381(1): 59-71, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19940980

RESUMO

The analgesic efficacy of opiates can be enhanced in inflammatory states due to peripheral and spinal alterations. We describe here that the analgesic effect induced by intrathecal (i.t.) morphine assessed by measuring thermal withdrawal latencies is enhanced in carrageenan-inflamed mice. The spinal micro-opioid receptor (MOR) population is not up-regulated as demonstrated by Western blot assays. In contrast, behavioural experiments show the involvement of changes in transduction mechanisms activated by spinal opioid receptors. The i.t. administration of the nitric oxide (NO) synthase inhibitor L-NMMA (3-30 microg) antagonised with a similar potency and efficacy morphine-induced analgesia in inflamed and non-inflamed mice, discarding that an increase in NO release could be responsible of the enhancement of morphine-induced analgesia. The analgesic effects evoked by the i.t. administration of the direct G(i/o) protein activator mastoparan (0.03-10 microg), but not those induced by the N-type calcium channel blocker omega-conotoxin GVIA (3-30 ng), were potentiated in inflamed mice, suggesting that postsynaptic and not presynaptic mechanisms could be involved. Furthermore, the inhibitory effects on morphine-induced analgesia produced by the G(i/o) protein inhibitor pertussis toxin (0.1-17 ng) or the G-coupled inwardly rectifying potassium (GIRK) channels inhibitor tertiapin-Q (0.75-750 ng) were greatly enhanced in inflamed mice. These results suggest that differences in the transduction mechanism activated by MOR at postsynaptic level, probably related with GIRK channels activity, could participate in the potentiation of morphine-induced spinal analgesia in acutely inflamed mice.


Assuntos
Analgésicos Opioides/administração & dosagem , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Morfina/administração & dosagem , Dor/metabolismo , Doença Aguda , Animais , Venenos de Abelha/farmacologia , Sinergismo Farmacológico , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Injeções Espinhais , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Peptídeos/administração & dosagem , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Venenos de Vespas/administração & dosagem
14.
Proc Natl Acad Sci U S A ; 106(38): 16451-6, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19805319

RESUMO

Peripheral interactions between nociceptive fibers and mast cells contribute to inflammatory pain, but little is known about mechanisms mediating neuro-immune communication. Here we show that metalloproteinase MT5-MMP (MMP-24) is an essential mediator of peripheral thermal nociception and inflammatory hyperalgesia. We report that MT5-MMP is expressed by CGRP-containing peptidergic nociceptors in dorsal root ganglia and that Mmp24-deficient mice display enhanced sensitivity to noxious thermal stimuli under basal conditions. Consistently, mutant peptidergic sensory neurons hyperinnervate the skin, a phenotype that correlates with changes in the regulated cleavage of the cell-cell adhesion molecule N-cadherin. In contrast to basal nociception, Mmp24(-/-) mice do not develop thermal hyperalgesia during inflammation, a phenotype that appears associated with alterations in N-cadherin-mediated cell-cell interactions between mast cells and sensory fibers. Collectively, our findings demonstrate an essential role of MT5-MMP in the development of dermal neuro-immune synapses and suggest that this metalloproteinase may be a target for pain control.


Assuntos
Gânglios Espinais/metabolismo , Hiperalgesia/fisiopatologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Nociceptores/metabolismo , Animais , Western Blotting , Células COS , Caderinas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Feminino , Imunofluorescência , Gânglios Espinais/citologia , Temperatura Alta , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/fisiopatologia , Masculino , Metaloproteinases da Matriz Associadas à Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/metabolismo , Doenças do Sistema Nervoso Periférico/fisiopatologia , Transfecção
15.
Neurosci Lett ; 465(3): 285-9, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19765404

RESUMO

Although previous studies describe the up-regulation of purinergic P2X(3) receptors expressed at peripheral nociceptive fibers in experimental painful neoplastic processes, the analgesic efficacy of P2X(3) receptor antagonists has not been tested in these settings. We study here the effect of the P2X(3) receptor antagonist, A-317491, on thermal hyperalgesia produced by the intratibial inoculation of NCTC 2472 fibrosarcoma cells to C3H/HeJ mice. The peritumoral administration of A-317491 (10-100 microg) dose-dependently attenuated osteosarcoma-induced thermal hyperalgesia without modifying thermal latencies measured in the contralateral paws. This antihyperalgesic effect was inhibited by the coadministration of naloxone-methiodide (0.1-1 microg) or the systemic injection of the selective mu-opioid receptor antagonist cyprodime (1 mg/kg), demonstrating the involvement of peripheral mu-opioid receptors. Furthermore, the antihyperalgesic effect induced by A-317491, was antagonised by the coadministration of an anti-enkephalin antibody supporting the participation of endogenous enkephalins. Consistent with this result, the antihyperalgesic effect induced by A-317491 was dramatically enhanced by the administration of an enkephalin-degrading inhibitor, Debio 0827, as demonstrated by isobolographic analysis. This synergism opens the theoretical possibility that the combination of both types of drugs could be useful to counteract some nociceptive symptoms derived from tumor development.


Assuntos
Encefalinas/metabolismo , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Osteossarcoma/complicações , Osteossarcoma/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Linhagem Celular Tumoral , Temperatura Alta , Masculino , Camundongos , Camundongos Endogâmicos C3H , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2X3
16.
Eur J Pharmacol ; 596(1-3): 50-5, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18692494

RESUMO

We have previously shown that stimulation of peripheral opioid receptors by exogenous opiates counteracts the thermal hyperalgesia elicited by a tibial osteosarcoma due to intraosteal inoculation of NCTC 2472 cells to mice. Aiming to study whether pheripheral endogenous enkephalins could also counteract this painful symptom, we assayed in this model the effects of PL37, an orally active dual inhibitor of enkephalin inactivating enzymes. Oral administration of PL37 (25 mg/kg) completely supressed osteosarcoma-induced thermal hyperalgesia through the activation of micro-opioid receptors, since the administration of cyprodime (1 mg/kg) inhibited its antihyperalgesic effect. Neither naltrindole (0.1 mg/kg) nor nor-binaltorphimine (10 mg/kg) modified this PL37-induced antihyperalgesic effect. Moreover, the inhibition of the antihyperalgesic effect induced by PL37 after the administration of naloxone-methiodide (2 mg/kg), a non selective opioid antagonist that does not cross the blood-brain barrier, demonstrates the involvement of peripheral opioid receptors. In contrast, centrally mediated effects may be detected when assaying a higher dose of PL37 (50 mg/kg). Besides, the administration of gabapentin (6.25-25 mg/kg, i.p.) dose-dependently inhibited osteosarcoma-induced thermal hyperalgesia. Interestingly, the combined administration of subeffective doses of PL37 and gabapentin completely prevented this type of thermal hyperalgesia. An isobolographic analysis of this interaction demonstrated a synergistic interaction between both drugs.


Assuntos
Aminas/uso terapêutico , Analgésicos/uso terapêutico , Neoplasias Ósseas/fisiopatologia , Ácidos Cicloexanocarboxílicos/uso terapêutico , Dissulfetos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Neprilisina/antagonistas & inibidores , Osteossarcoma/fisiopatologia , Propilaminas/uso terapêutico , Ácido gama-Aminobutírico/uso terapêutico , Administração Oral , Animais , Neoplasias Ósseas/complicações , Sinergismo Farmacológico , Gabapentina , Temperatura Alta , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Camundongos , Naloxona/análogos & derivados , Naloxona/farmacologia , Osteossarcoma/complicações , Compostos de Amônio Quaternário/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/fisiologia
17.
Cell Mol Neurobiol ; 28(7): 981-90, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18360770

RESUMO

The stimulation of peripheral opioid receptors counteracts thermal hyperalgesia produced by the intratibial inoculation of NCTC 2472 cells in mice, through the activation of the nitric oxide/cGMP/ATP-sensitive K+-channels (NO/cGMP/K(+) (ATP)) cascade (Menéndez et al. 2007, Neuropharmacology 53:71-80). We aimed to elucidate whether this peripheral opioid antihyperalgesic effect is exclusive to this model or might also occur in other types of bone neoplastic processes. In C57BL/6 mice intratibially inoculated with B16-F10 melanoma cells, the progressive tumoral damage was accompanied by the establishment of thermal hyperalgesia (unilateral hot plate test) and mechanical allodynia (von Frey test). Intraplantar administration of loperamide (15 microg, 30 min before) inhibited thermal hyperalgesia, but did not modify the intense mechanical allodynia. The fact that the coadministration of naloxone-methiodide (5 microg) completely suppressed the thermal antihyperalgesic effect induced by loperamide indicates its production through the stimulation of peripheral opioid receptors. Furthermore, its prevention by the coadministration of the non-selective inhibitor of the NO synthase, N(G)-monomethyl-L-arginine (L-NMMA, 10 microg), the selective inhibitor of neural NOS, N-omega-propyl-L-arginine (1-10 microg), or the K+ (ATP) channel blocker, glibenclamide (10 microg) demonstrated the involvement of the NO/cGMP/K(+) (ATP) pathway in the antihyperalgesic effect induced by loperamide. Overall, the present results show that the intratibial inoculation of B16-F10 cells to C57BL/6 mice evokes thermal hyperalgesia and mechanical allodynia and that, as occurred in the osteosarcoma model, the stimulation of peripheral opioid receptors is not effective in modifying neoplastic allodynia but completely inhibits thermal hyperalgesia through the activation of the NO/cGMP/K+ (ATP) cascade.


Assuntos
Neoplasias Ósseas/complicações , Osso e Ossos/inervação , Hiperalgesia/tratamento farmacológico , Loperamida/farmacologia , Nociceptores/efeitos dos fármacos , Receptores Opioides/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Osso e Ossos/fisiopatologia , Linhagem Celular Tumoral , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Hiperalgesia/etiologia , Hiperalgesia/fisiopatologia , Canais KATP/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Nociceptores/metabolismo , Medição da Dor , Bloqueadores dos Canais de Potássio/farmacologia , Receptores Opioides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tíbia/inervação , Tíbia/fisiopatologia , Tíbia/cirurgia , Transplante de Tecidos/métodos
18.
Life Sci ; 81(8): 673-82, 2007 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-17692876

RESUMO

Based on the well established involvement of IL-1beta in inflammatory hyperalgesia, we have assessed the possible role played by IL-1beta in a murine model of bone cancer-induced pain. With this aim, we measured IL-1beta levels at the region of the tibia and the spinal cord in mice bearing a tibial osteosarcoma induced by the inoculation of NCTC 2472 cells, and we tested whether the IL-1 receptor antagonist, anakinra, inhibits some hypernociceptive reactions evoked by the neoplastic injury. Parallel experiments were performed in mice with a chronic inflammatory process (intraplantar injection of complete Freund's adjuvant, CFA). IL-1beta levels were increased in the tibial region of osteosarcoma-bearing mice and in the paws of inflamed mice. To a lesser extent, the content of IL-1beta in the spinal cord was also augmented in both situations. Osteosarcoma-induced thermal hyperalgesia was inhibited by 30 and 100 mg/kg of systemic anakinra, but only 300 mg/kg prevented inflammatory thermal hyperalgesia. Mechanical hyperalgesia induced by the osteosarcoma was blocked by 100 and 300 mg/kg of anakinra, whereas a partial reversion of inflammatory mechanical hyperalgesia was induced by 300 mg/kg. Anakinra, intrathecally administered (1 and 10 microg) did not modify hyperalgesia of either origin. Besides, both tumoral and inflammatory mechanical allodynia remained unaltered after the administration of anakinra. In conclusion, some hyperalgesic symptoms observed in this model of bone cancer are mediated by the peripheral release of IL-1beta and may be inhibited by antagonists of type I IL-1 receptors with a similar or greater potency than symptoms produced by inflammation.


Assuntos
Neoplasias Ósseas/complicações , Hiperalgesia/tratamento farmacológico , Inflamação/complicações , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Interleucina-1beta/metabolismo , Osteossarcoma/complicações , Receptores de Interleucina-1/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund , Hiperalgesia/etiologia , Hiperalgesia/psicologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Injeções Espinhais , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Camundongos , Camundongos Endogâmicos C3H , Osteossarcoma/metabolismo , Medição da Dor/efeitos dos fármacos , Estimulação Física , Pressão , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
19.
Neuropharmacology ; 53(1): 71-80, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17543351

RESUMO

Experiments were designed to elucidate the involvement of nitric oxide (NO) in the antihyperalgesic effect induced by the activation of peripheral mu-opioid receptors on osteosarcoma-induced thermal hyperalgesia in mice. Since this pathway has previously been shown to be involved in the antihyperalgesic effect induced by some drugs--including opiates--on inflammatory pain, experiments were also performed in inflamed mice. The intraplantar administration of loperamide (15 microg) abolishes the thermal hyperalgesia that appears 4 weeks after the intratibial inoculation of NCTC 2472 cells in C3H/HeJ mice. The blockade of this effect by coadministering a peripheral opioid receptor antagonist (naloxone methiodide), a nitric oxide synthase (NOS) inhibitor (L-NMMA), a soluble guanylyl cyclase inhibitor (ODQ), a PKG inhibitor (KT-5823) or a K(+)(ATP)-channel blocker (glibenclamide) shows the involvement of a NO/cGMP/K(+)(ATP)-channel pathway. Accordingly the administration of loperamide produced, in osteosarcoma-bearing mice, an increase in the concentrations of NO metabolites, nitrites and nitrates, extracted from paws. The selective inhibitor of eNOS L-NIO, but not the inhibitors of nNOS (N-omega-propyl-L-arginine) or iNOS (1400w), blocked the effect of loperamide on osteosarcoma-induced hyperalgesia and also the endogenous opioid peripheral hypoalgesia that appears during the initial stages of the development of this osteosarcoma. Although this pathway also participates in the inhibitory effect of loperamide on the thermal hyperalgesia induced by administration of complete Freund's adjuvant, only selective inhibitors of nNOS or iNOS antagonized this effect. Our results demonstrate that the activation of a NO/cGMP/K(+)(ATP)-channel triggered by eNOS participates in the peripheral antihyperalgesic of loperamide on osteosarcoma-induced thermal hyperalgesia.


Assuntos
Neoplasias Ósseas/complicações , Hiperalgesia/enzimologia , Hiperalgesia/etiologia , Óxido Nítrico/fisiologia , Receptores Opioides/fisiologia , Análise de Variância , Animais , Carbazóis/administração & dosagem , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Glibureto/administração & dosagem , Hiperalgesia/tratamento farmacológico , Indóis/administração & dosagem , Loperamida/uso terapêutico , Camundongos , Camundongos Endogâmicos C3H , NG-Nitroarginina Metil Éster/administração & dosagem , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Nitratos/metabolismo , Nitritos/metabolismo , Medição da Dor
20.
Peptides ; 27(11): 2778-85, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16930772

RESUMO

Transient thermal, but not mechanical, hypoalgesia appears at the early stages of the development of an hyperalgesic murine osteosarcoma. This hypoalgesia is suppressed by the administration of naloxone, its peripherally acting analog naloxone methiodide, the mu- and delta-opioid receptor antagonists cyprodime and naltrindole, or the CRF receptor antagonist, alpha-helical CRF (9-41). When immunohistochemical assays were performed with an anti-beta-endorphin antibody, whose in vivo administration suppressed the analgesia, labeled mononuclear immune cells appeared both inside and surrounding the tumoral tissue. In conclusion, the peripheral action of beta-endorphin, released in response to the osteosarcoma seems responsible for the observed thermal analgesia.


Assuntos
Hiperalgesia/fisiopatologia , Hipestesia/fisiopatologia , Osteossarcoma/fisiopatologia , beta-Endorfina/metabolismo , Animais , Células Cultivadas , Hiperalgesia/etiologia , Hipestesia/etiologia , Camundongos , Neurotransmissores/metabolismo , Osteossarcoma/complicações , Receptores Opioides/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA