Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Bioorg Chem ; 115: 105231, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34388485

RESUMEN

The analgesic peptide DD04107 (Pal-EEMQRR-NH2) and its acetylated analogue inhibit α-calcitonin gene-related peptide (α-CGRP) exocytotic release from primary sensory neurons. Examining the crystal structure of the SNARE-Synaptotagmin-1(Syt1) complex, we hypothesized that these peptides could inhibit neuronal exocytosis by binding to Syt1, hampering at least partially its interaction with the SNARE complex. To address this hypothesis, we first interrogate the role of individual side-chains on the inhibition of α-CGRP release, finding that E1, M3, Q4 and R6 residues were crucial for activity. CD and NMR conformational analysis showed that linear peptides have tendency to adopt α-helical conformations, but the results with cyclic analogues indicated that this secondary structure is not needed for activity. Isothermal titration calorimetry (ITC) measurements demonstrate a direct interaction of some of these peptides with Syt1-C2B domain, but not with Syt7-C2B region, indicating selectivity. As expected for a compound able to inhibit α-CGRP release, cyclic peptide derivative Pal-E-cyclo[EMQK]R-NH2 showed potent in vivo analgesic activity, in a model of inflammatory pain. Molecular dynamics simulations provided a model consistent with KD values for the interaction of peptides with Syt1-C2B domain, and with their biological activity. Altogether, these results identify Syt1 as a potential new analgesic target.


Asunto(s)
Analgésicos/farmacología , Lipopéptidos/farmacología , Dolor/tratamiento farmacológico , Sinaptotagmina I/antagonistas & inhibidores , Analgésicos/síntesis química , Analgésicos/química , Animales , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Péptido Relacionado con Gen de Calcitonina/metabolismo , Relación Dosis-Respuesta a Droga , Exocitosis/efectos de los fármacos , Lipopéptidos/síntesis química , Lipopéptidos/química , Masculino , Ratones , Simulación de Dinámica Molecular , Estructura Molecular , Dolor/metabolismo , Relación Estructura-Actividad , Sinaptotagmina I/metabolismo
2.
Cell Mol Neurobiol ; 39(8): 1115-1124, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31203533

RESUMEN

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.


Asunto(s)
Analgesia , Quimiocina CCL1/administración & dosificación , Endocannabinoides/metabolismo , Temperatura , Analgésicos/farmacología , Animales , Ácidos Araquidónicos/metabolismo , Ciclofosfamida , Glicéridos/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/metabolismo , Masculino , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Receptores CCR8/metabolismo
3.
Brain Behav Immun ; 62: 151-161, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28126501

RESUMEN

We show here that the intraplantar administration of CCL5 in mice produces hyperalgesia at low doses but activates compensatory antinociceptive mechanisms at doses slightly higher. Thus, the injection of 3-10ng of CCL5 evoked thermal hyperalgesia through the activation of CCR1 and CCR5 receptors, as demonstrated by the inhibitory effect exerted by the selective antagonists J113863 (0.01-0.1µg) and DAPTA (0.3-3µg), respectively. The prevention of this hyperalgesia by diclofenac (1-10µg), the inhibitors of COX-1 SC-560 (0.1-1µg) or COX-2 celecoxib (1-5µg), the TRPV1 antagonist capsazepine (0.03-0.3µg) or the TRPA1 antagonist HC030031 (10-50µg) demonstrates the involvement of prostaglandin synthesis and TRP sensitization in CCL5-evoked hyperalgesia. Doses of CCL5 higher than 17µg did not evoke hyperalgesia. However, this effect was restored by the administration of naloxone-methiodide (5µg), nor-binaltorphimine (10mg/kg) or an anti-dynorphin A antibody (0.62-2.5ng). The administration of 30ng of CCL5 also induced hyperalgesia in mice with reduced number of circulating white blood cells in response to cyclophosphamide or with selective neutrophil depletion induced by an anti-Ly6G antibody. In fact, the number of neutrophils present in paws treated with 30ng of CCL5 was greater than in paws receiving the administration of the hyperalgesic dose of 10ng. Finally, the expression of the endogenous opioid peptide dynorphin A was demonstrated by double immunofluorescence assays in these neutrophils attracted by CCL5. These results support previous data describing the hyperalgesic properties of CCL5 and constitute the first indication that a chemokine of the CC group can activate endogenous analgesic mechanisms.


Asunto(s)
Quimiocina CCL5 , Hiperalgesia/inducido químicamente , Receptores CCR1/metabolismo , Receptores CCR5/metabolismo , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Celecoxib/administración & dosificación , Celecoxib/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Diclofenaco/administración & dosificación , Diclofenaco/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Masculino , Ratones , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos
4.
Prostate ; 75(1): 70-83, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25263196

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/secundario , Hiperalgesia/fisiopatología , Dolor Nociceptivo/fisiopatología , Neoplasias de la Próstata/patología , Tibia/patología , Animales , Proteínas de Unión al Calcio , Línea Celular Tumoral , Quimiocinas/metabolismo , Difosfonatos/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Proteína Ácida Fibrilar de la Glía , Hiperalgesia/tratamiento farmacológico , Imidazoles/farmacología , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Morfina/farmacología , Trasplante de Neoplasias , Proteínas del Tejido Nervioso , Dolor Nociceptivo/tratamiento farmacológico , Receptores CCR2/metabolismo , Médula Espinal/patología , Ácido Zoledrónico
5.
Cell Mol Neurobiol ; 34(1): 143-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24122510

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Óseas/complicaciones , Quimiocina CCL2/metabolismo , Hiperalgesia/etiología , Microglía/metabolismo , Osteosarcoma/complicaciones , Médula Espinal/metabolismo , Animales , Astrocitos/efectos de los fármacos , Benzoxazinas/farmacología , Western Blotting , Línea Celular Tumoral , Quimiocina CCL2/genética , Ensayo de Inmunoadsorción Enzimática , Ganglios Espinales/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/metabolismo , Vértebras Lumbares/patología , Ratones , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Compuestos de Espiro/farmacología , Regulación hacia Arriba/efectos de los fármacos
6.
J Neuroimmune Pharmacol ; 19(1): 44, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39152360

RESUMEN

The systemic administration of interleukin-16 (IL-16, 3-30 ng/kg) induced thermal hyperalgesia in mice, that was prevented by the acute injection of an anti-CD4 antibody (1 µg/kg), the depletion of circulating white blood cells by cyclophosphamide or the specific reduction of circulating CD4+ cells provoked by a high dose of an anti-CD4 antibody (30 µg/mouse, 24 h before). IL-16-induced hyperalgesia was locally inhibited after intraplantar (i.pl.) administration of the non-selective cyclooxygenase (COX) inhibitor diclofenac, the COX-1 inhibitor SC-560, the COX-2 inhibitor celecoxib, the TRPV1 antagonist capsazepine or the TRPA1 antagonist HC030031, thus demonstrating that prostaglandins and TRP channels are involved in this effect. The i.pl. administration of low doses of IL-16 (0.1-1 ng) evoked local hyperalgesia suggesting the possibility that IL-16 could participate in hypernociception associated to local tissue injury. Accordingly, IL-16 concentration measured by ELISA was increased in paws acutely inflamed with carrageenan or chronically inflamed with complete Freund´s adjuvant (CFA). This augmentation was reduced after white cell depletion with cyclophosphamide or neutrophil depletion with an anti-Ly6G antibody. Immunofluorescence and flow cytometry experiments showed that the increased concentration of IL-16 levels found in acutely inflamed paws is mainly related to the infiltration of IL-16+ neutrophils, although a reduced number of IL-16+ lymphocytes was also detected in paws inflamed with CFA. Supporting the functional role of IL-16 in inflammatory hypernociception, the administration of an anti-IL-16 antibody dose-dependently reduced carrageenan- and CFA-induced thermal hyperalgesia and mechanical allodynia. The interest of IL-16 as a target to counteract inflammatory pain is suggested.


Asunto(s)
Hiperalgesia , Inflamación , Interleucina-16 , Animales , Ratones , Hiperalgesia/tratamiento farmacológico , Masculino , Inflamación/tratamiento farmacológico , Inflamación/inducido químicamente , Interleucina-16/metabolismo
7.
J Neurosci Methods ; 386: 109783, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36610617

RESUMEN

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.


Asunto(s)
Hiperalgesia , Dolor , Animales , Dimensión del Dolor , Analgésicos/farmacología
8.
J Pharmacol Exp Ther ; 341(3): 634-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22393248

RESUMEN

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.


Asunto(s)
Analgésicos/farmacología , Modelos Animales de Enfermedad , Exocitosis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lipopéptidos/farmacología , Neuralgia/tratamiento farmacológico , Neuronas/efectos de los fármacos , Analgésicos/efectos adversos , Analgésicos/farmacocinética , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Carragenina/toxicidad , Relación Dosis-Respuesta a Droga , Hiperalgesia/tratamiento farmacológico , Inyecciones Subcutáneas , Lipopéptidos/efectos adversos , Lipopéptidos/farmacocinética , Masculino , Ratones , Ratones Endogámicos C3H , Neoplasias Experimentales/patología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Factores de Tiempo
10.
Proc Natl Acad Sci U S A ; 106(38): 16451-6, 2009 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-19805319

RESUMEN

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.


Asunto(s)
Ganglios Espinales/metabolismo , Hiperalgesia/fisiopatología , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Nociceptores/metabolismo , Animales , Western Blotting , Células COS , Cadherinas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Femenino , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/citología , Calor , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Metaloproteinasas de la Matriz Asociadas a la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Transfección
11.
Life Sci ; 291: 120302, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34999112

RESUMEN

AIMS: To explore the mechanisms involved in the transformation of analgesia produced by low doses of CCL4 (pg/kg) to hyperalgesia when higher doses (ng/kg) are administered to mice. MAIN METHODS: The unilateral hot plate test was used to assess thermal nociception. CD3+, CD4+ or CD8+ blood cells were depleted with selective antibodies. Expression of CCR5 and IL-16 in lymphocytes was studied by flow cytometry and IL-16 blood levels were measured by ELISA. IL-16 and CD8 were detected by immunofluorescence. KEY FINDINGS: IL-16 and CCR5 expression were demonstrated in CD4+ and CD8+ T-lymphocytes by flow cytometry. Furthermore, CCL4-induced hyperalgesia was abolished by reducing circulating T-lymphocyte levels or by selectively depleting CD4+ lymphocytes. In contrast, when the anti-CD4 antibody was acutely administered, CCL4 induced analgesia instead of hyperalgesia. A similar response was obtained when administering A-770041, that prevents CD4-mediated CCR5 desensitization by inhibiting p56lck kinase. As occurred with the analgesic effect evoked by low doses of CCL4, analgesia evoked by combining CCL4 and A-770041 was reverted by naloxone, naltrindole or an anti-met-enk antibody. Interestingly, flow cytometry assays showed that the number of CD8+, but not CD4+, T-cells expressing IL-16 is reduced after the acute administration of CCL4, a result compatible with the description that CD8+-lymphocytes can rapidly release preformed IL-16. Accordingly, the rise in IL-16 blood concentration evoked by CCL4 was prevented after CD8+ lymphocyte depletion. SIGNIFICANCE: CCL4-evoked hyperalgesia is related to the desensitization of CCR5 in CD4+ T-cells and to the release of IL-16 from CD8+ lymphocytes.


Asunto(s)
Quimiocina CCL4/metabolismo , Hiperalgesia/metabolismo , Nocicepción/efectos de los fármacos , Analgesia/métodos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL4/inmunología , Quimiocina CCL4/farmacología , Citometría de Flujo/métodos , Calor , Masculino , Ratones , Naloxona/farmacología , Dolor/metabolismo , Receptores CCR5/metabolismo
12.
Fundam Clin Pharmacol ; 35(6): 1109-1118, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33905573

RESUMEN

BACKGROUND: The chemokine CC motif ligand 1 (CCL1) participates in immune cell recruitment and, as other chemokines, is also involved in nociceptive processing. In contrast with previous reports indicating its participation in allodynia and cold hypernociception when spinally administered, its ability to evoke heat thermal analgesia, mediated by circulating leukocytes and endocannabinoids, after systemic administration has recently been reported. OBJECTIVES: Aiming to explore the role played by CCL1 on spinal nociception, we study here the effect of its intrathecal administration on thermal nociception in mice. METHODS: Behavioral nociceptive assays, immunohistochemical experiments, white cell blood depletion procedures and qRT-PCR experiments were performed. RESULTS: The intrathecal administration of CCL1 (0.3-30 ng) produced analgesia as measured by the unilateral hot plate test. This effect peaked 1 h after injection, was prevented by the CCR8 antagonist R243 and was accompanied by a reduction of c-Fos expression in spinal neurons. Whereas blood leukocyte depletion did not modify it, analgesia was abolished by the microglial inhibitor minocycline, but not the astroglial inhibitor aminoadipate. Furthermore, antinociception remained unmodified by the coadministration of cannabinoid type 1 or 2 receptors antagonists. However, it was reversed by naloxone but not by selective blockade of mu- or delta-opioid receptors. The inhibitory effect induced by the selective kappa-opioid receptor antagonist, nor-binaltorphimine, and by an anti-dynorphin A 1-17 antibody indicates that analgesia evoked by spinal CCL1 is mediated by endogenous dynorphins acting on kappa-opioid receptors. CONCLUSIONS: Endogenous dynorphin and microglia behave as key players in heat thermal analgesia evoked by spinal CCL1 in mice.


Asunto(s)
Analgesia , Receptores Opioides kappa , Animales , Quimiocina CCL1 , Ligandos , Ratones , Morfina , Antagonistas de Narcóticos/farmacología , Médula Espinal
13.
Cell Mol Neurobiol ; 30(1): 113-21, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19655242

RESUMEN

The analgesic effect induced by opiates is often potentiated during experimental inflammatory processes. We describe here that lower doses of systemic morphine are necessary to increase thermal withdrawal latencies measured in both hind paws of mice acutely inflamed with carrageenan than in healthy ones. This bilateral potentiation seems mediated through spinal opioid receptors since it is inhibited by the intrathecal (i.t.), but not intraplantar (i.pl.) administration of the opioid receptor antagonist naloxone-methiodide, and also appears when morphine is i.t. administered. Furthermore, the i.pl. administration of the nitric oxide (NO) synthase inhibitor, L-NMMA, or the K (ATP) (+) -channel blocker, glibenclamide, to carrageenan-inflamed mice inhibits the enhanced effect of systemic morphine in the paw that receives the injection of the drug, without affecting the potentiation observed in the contralateral one. The i.pl. administration of L-NMMA also partially antagonised the analgesic effect induced by i.t. morphine in inflamed mice. Finally, the increased analgesic effect evoked by the i.pl. administration of the NO donor SIN-1 either in the inflamed or in the contralateral paw of carrageenan-inflamed mice suggests that enhanced responsiveness to the peripheral analgesic effect of NO may be also underlying the bilateral potentiation of morphine-induced analgesia in acutely inflamed mice.


Asunto(s)
Analgesia , Inflamación/patología , Morfina/farmacología , Médula Espinal/patología , Enfermedad Aguda , Animales , Carragenina , Gliburida/administración & dosificación , Gliburida/farmacología , Inflamación/inducido químicamente , Inyecciones Espinales , Ratones , Molsidomina/administración & dosificación , Molsidomina/análogos & derivados , Molsidomina/farmacología , Morfina/administración & dosificación , Naloxona/administración & dosificación , Naloxona/análogos & derivados , Naloxona/farmacología , Nociceptores/metabolismo , Dolor/patología , Compuestos de Amonio Cuaternario/administración & dosificación , Compuestos de Amonio Cuaternario/farmacología , Temperatura , omega-N-Metilarginina/administración & dosificación , omega-N-Metilarginina/farmacología
14.
Biochem Pharmacol ; 175: 113903, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32156658

RESUMEN

As recently described, the administration of extremely low doses (pg/kg) of CCL4 (Macrophage inflammatory protein 1ß, MIP-1ß) can induce antinociceptive effects in mice (García-Domínguez et al., 2019b). We describe here that hydrodynamic delivery of a plasmid containing CCL4 cDNA provokes a biphasic response consisting in an initial thermal hyperalgesic reaction for 8 days followed by analgesia at days 10-12, being both responses blocked after the administration of the CCR5 antagonist DAPTA. Both the luminiscence evoked in liver after the administration of a plasmid containing CCL4 and luciferase cDNAs and the hepatic concentration of CCL4 measured by ELISA were maximal 4 days after plasmid administration and markedly diminished at day 10. A dose-effect curve including a wide dose range of exogenous CCL4 revealed thermal analgesia after the administration of 10-100 pg/kg whereas 1000 times higher doses (30-100 ng/kg) induced, instead, thermal hyperalgesia inhibited by DAPTA. This hyperalgesia was absent in mice with reduced white blood cells after cyclophosphamide treatment, thus supporting the involvement of circulating leukocytes. A multiarray bioluminescent assay revealed increased plasma levels of IL-1α, CCL2, CXCL1, CXCL13, IL-16 and TIMP-1 in mice treated with 100 ng/kg of CCL4. The hyperalgesic response evoked by CCL4 was prevented by IL-1R, CXCR2 or CCR2 antagonists or by the neutralization of CXCL13 or IL-16, but not TIMP-1, with selective antibodies. The administration of the anti-IL-16 antibody was the unique treatment able to convert hyperalgesia evoked by 100 ng/kg of CCL4 in an analgesic effect. The ability of IL-16 to evoke hypernociception was confirmed by studying the response to its exogenous administration (10-30 ng/kg). In summary, the present results demonstrate that CCL4 induces a dual modulation of nociception and describe some mechanisms involved in the hyperalgesic response evoked by this chemokine.


Asunto(s)
Quimiocina CCL4/administración & dosificación , Técnicas de Transferencia de Gen , Calor/efectos adversos , Hiperalgesia/tratamiento farmacológico , Nocicepción/fisiología , Animales , Quimiocina CCL4/genética , Relación Dosis-Respuesta a Droga , Hiperalgesia/genética , Masculino , Ratones , Nocicepción/efectos de los fármacos
15.
Sci Rep ; 10(1): 18599, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33122720

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/farmacología , Dolor en Cáncer/tratamiento farmacológico , Piperidinas/farmacología , Receptores Opioides mu/metabolismo , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/metabolismo , Dolor en Cáncer/metabolismo , Línea Celular Tumoral , Fentanilo/farmacología , Concentración de Iones de Hidrógeno , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Ligandos , Masculino , Melanoma Experimental/complicaciones , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Morfinanos/farmacología , Naloxona/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología
16.
Mol Neurobiol ; 56(3): 1578-1595, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29907903

RESUMEN

In the present study, we characterize the antinociceptive effects produced by the chemokine CCL4 in mice. The intraplantar administration of very low doses of CCL4 (0.1-3 pg) produced bilateral antinociception assessed by the unilateral hot-plate test (UHP) without evoking chemotactic responses at the injection site. Moreover, the subcutaneous administration of CCL4 (3-100 pg/kg) also yielded bilateral antinociception in the UHP and the paw pressure test and reduced the number of spinal neurons that express Fos protein in response to noxious stimulation. The implication of peripheral CCR5 but not CCR1 in CCL4-evoked antinociception was deduced from the inhibition produced by systemic but not intrathecal, administration of the CCR5 antagonist DAPTA, and the inefficacy of the CCR1 antagonist J113863. Besides, the inhibition observed after subcutaneous but not intrathecal administration of naloxone demonstrated the involvement of peripheral opioids and the efficacy of naltrindole but not cyprodime or nor-binaltorphimine supported the participation of δ-opioid receptors. In accordance, plasma levels of met-enkephalin, but not ß-endorphin, were augmented in response to CCL4. Likewise, CCL4-evoked antinociception was blocked by the administration of an anti-met-enk antibody. Leukocyte depletion experiments performed with cyclophosphamide, anti-Ly6G, or anti-CD3 antibodies indicated that the antinociceptive effect evoked by CCL4 depends on circulating T lymphocytes. Double immunofluorescence experiments showed a four times more frequent expression of met-enk in CD4+ than in CD8+ T lymphocytes. CCL4-induced antinociception almost disappeared upon CD4+, but not CD8+, lymphocyte depletion with selective antibodies, thus supporting that the release of met-enk from CD4+ lymphocytes underlies the opioid antinociceptive response evoked by CCL4.


Asunto(s)
Analgésicos/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Quimiocina CCL4/uso terapéutico , Encefalina Metionina/metabolismo , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Analgésicos/farmacología , Animales , Linfocitos T CD4-Positivos/metabolismo , Quimiocina CCL4/farmacología , Ratones , Naloxona/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Dolor/metabolismo , Dimensión del Dolor
17.
Cell Mol Neurobiol ; 28(7): 981-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18360770

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/complicaciones , Huesos/inervación , Hiperalgesia/tratamiento farmacológico , Loperamida/farmacología , Nociceptores/efectos de los fármacos , Receptores Opioides/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Huesos/fisiopatología , Línea Celular Tumoral , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Canales KATP/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Nociceptores/metabolismo , Dimensión del Dolor , Bloqueadores de los Canales de Potasio/farmacología , Receptores Opioides/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tibia/inervación , Tibia/fisiopatología , Tibia/cirugía , Trasplante de Tejidos/métodos
18.
Eur J Pharmacol ; 596(1-3): 50-5, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18692494

RESUMEN

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.


Asunto(s)
Aminas/uso terapéutico , Analgésicos/uso terapéutico , Neoplasias Óseas/fisiopatología , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Disulfuros/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Neprilisina/antagonistas & inhibidores , Osteosarcoma/fisiopatología , Propilaminas/uso terapéutico , Ácido gamma-Aminobutírico/uso terapéutico , Administración Oral , Animales , Neoplasias Óseas/complicaciones , Sinergismo Farmacológico , Gabapentina , Calor , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Ratones , Naloxona/análogos & derivados , Naloxona/farmacología , Osteosarcoma/complicaciones , Compuestos de Amonio Cuaternario/farmacología , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/fisiología , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/fisiología , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/fisiología
19.
Neuropharmacology ; 53(1): 71-80, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17543351

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/complicaciones , Hiperalgesia/enzimología , Hiperalgesia/etiología , Óxido Nítrico/fisiología , Receptores Opioides/fisiología , Análisis de Varianza , Animales , Carbazoles/administración & dosificación , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Gliburida/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Indoles/administración & dosificación , Loperamida/uso terapéutico , Ratones , Ratones Endogámicos C3H , NG-Nitroarginina Metil Éster/administración & dosificación , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Nitratos/metabolismo , Nitritos/metabolismo , Dimensión del Dolor
20.
Life Sci ; 81(8): 673-82, 2007 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-17692876

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/complicaciones , Hiperalgesia/tratamiento farmacológico , Inflamación/complicaciones , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1beta/metabolismo , Osteosarcoma/complicaciones , Receptores de Interleucina-1/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Neoplasias Óseas/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Adyuvante de Freund , Hiperalgesia/etiología , Hiperalgesia/psicología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inyecciones Espinales , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Ratones , Ratones Endogámicos C3H , Osteosarcoma/metabolismo , Dimensión del Dolor/efectos de los fármacos , Estimulación Física , Presión , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA