Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.450
Filtrar
1.
Neuropharmacology ; 252: 109947, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38631564

RESUMEN

A growing body of research indicates that ß-caryophyllene (BCP), a constituent present in a large number of plants, possesses significant therapeutic properties against CNS disorders, including alcohol and psychostimulant use disorders. However, it is unknown whether BCP has similar therapeutic potential for opioid use disorders. In this study, we found that systemic administration of BCP dose-dependently reduced heroin self-administration in rats under an FR2 schedule of reinforcement and partially blocked heroin-enhanced brain stimulation reward in DAT-cre mice, maintained by optical stimulation of midbrain dopamine neurons at high frequencies. Acute administration of BCP failed to block heroin conditioned place preference (CPP) in male mice, but attenuated heroin-induced CPP in females. Furthermore, repeated dosing with BCP for 5 days facilitated the extinction of CPP in female but not male mice. In the hot plate assay, pretreatment with the same doses of BCP failed to enhance or prolong opioid antinociception. Lastly, in a substitution test, BCP replacement for heroin failed to maintain intravenous BCP self-administration, suggesting that BCP itself has no reinforcing properties. These findings suggest that BCP may have certain therapeutic effects against opioid use disorders with fewer unwanted side-effects by itself.


Asunto(s)
Heroína , Sesquiterpenos Policíclicos , Autoadministración , Animales , Masculino , Heroína/administración & dosificación , Sesquiterpenos Policíclicos/farmacología , Sesquiterpenos Policíclicos/administración & dosificación , Femenino , Ratones , Ratas , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Sesquiterpenos/farmacología , Sesquiterpenos/administración & dosificación , Ratas Sprague-Dawley , Relación Dosis-Respuesta a Droga , Condicionamiento Operante/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Refuerzo en Psicología , Recompensa , Ratones Transgénicos , Nocicepción/efectos de los fármacos , Ratones Endogámicos C57BL
2.
Biomed Pharmacother ; 174: 116544, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38599058

RESUMEN

The current study was designed to investigate the potential of a synthetic therapeutic agent for better management of pain and inflammation, exhibiting minimal to non-existent ulcerogenic effects. The effect of 1-(2-chlorobenzoyl)-3-(2,3-dichlorophenyl) thiourea was assessed through model systems of nociception and anti-inflammatory activities in mice. In addition, the ulcerogenic potential was evaluated in rats using the NSAID-induced pyloric ligation model, followed by histopathological and biochemical analysis. The test was conducted on eight groups of albino rats, comprising of group I (normal saline), groups II and III (aspirin® at doses of 100 mg/kg and 150 mg/kg, respectively), groups IV and V (indomethacin at doses of 100 mg/kg and 150 mg/kg, respectively), and groups VI, VII, and VIII (lead-compound at 15 mg/kg, 30 mg/kg and 45 mg/kg doses, respectively). Furthermore, molecular docking analyses were performed to predict potential molecular target site interactions. The results showed that the lead-compound, administered at doses of 15, 30, and 45 mg/kg, yielded significant reductions in chemically and thermally induced nociceptive pain, aligning with the levels observed for aspirin® and tramadol. The compound also effectively suppressed inflammatory response in the carrageenan-induced paw edema model. As for the ulcerogenic effects, the compound groups displayed no considerable alterations compared to the aspirin® and indomethacin groups, which displayed substantial increases in ulcer scores, total acidity, free acidity, and gastric juice volume, and a decrease in gastric juice pH. In conclusion, these findings suggest that our test compound exhibits potent antinociceptive, anti-inflammatory properties and is devoid of ulcerogenic effects.


Asunto(s)
Inflamación , Simulación del Acoplamiento Molecular , Nocicepción , Úlcera Gástrica , Tiourea , Animales , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología , Úlcera Gástrica/tratamiento farmacológico , Tiourea/análogos & derivados , Tiourea/farmacología , Masculino , Nocicepción/efectos de los fármacos , Ratones , Inflamación/tratamiento farmacológico , Inflamación/patología , Ratas , Ratas Wistar , Analgésicos/farmacología , Analgésicos/química , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/química , Simulación por Computador , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Mucosa Gástrica/metabolismo , Indometacina/farmacología , Dolor/tratamiento farmacológico , Dolor/inducido químicamente , Dolor/patología , Antiinflamatorios/farmacología
3.
J Ethnopharmacol ; 290: 115088, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35149131

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ben-Cha-Moon-Yai (BMY) remedy used in Thai traditional medicine as an anti-inflammatory, analgesic, and antipyretic agent compromises five herbal root extracts of equal weights: Aegle marmelos (L.) Corrêa (AM), Oroxylum indicum (L.) Kurz (OI), Dimocarpus longan Lour. (DL), Dolichandrone serrulata (Wall. ex DC.) Seem. (DS), and Walsura trichostemon Miq. (WT). AIM OF THE STUDY: To assess the anti-nociceptive and anti-inflammatory effects of the root extracts of all five species of BMY in experimental animal (mouse) models to ensure the rational use of herbal products in Thai traditional medicine. MATERIALS AND METHODS: Root extracts prepared by ethanol and water extraction were used for the biological assays in animal models at five dose levels: 25, 50,100,200 & 400 mg/kg. The anti-nociceptive activity was evaluated based on hot-plate latency, duration of paw licking induced by formalin, and abdominal writhing induced by acetic acid. Carrageenan- and prostaglandin-induced paw oedema models were used to determine the anti-inflammatory activity. RESULTS: The oral administration of AM, DS and WT root extracts displayed significant analgesic effects in the hot-plate test, both phases (early and late) of formalin test and acetic-acid induced writhing test at different dose levels. OI and DL only produced significant analgesia in the late phase of the formalin test and writhing test. The pretreatment of animals with the non-selective opioid receptor antagonist naloxone, reverse AM, DS and WT induced-antinociceptive activity. In both carrageenan and prostaglandin-induced paw oedema tests, all five herbal plant root extracts significantly reduced paw oedema at 3 h or more at different dose levels. Rotarod test results showed no effects of five herbal plant root extracts on the balance and the motor coordination at the highest dose level evaluated (400 mg/kg). CONCLUSION: The root extracts of AM, DS, and WT possess both central and peripheral anti-nociceptive properties, while OI and DL possess only peripheral analgesic properties. All five root extracts own anti-inflammatory properties, which might be due to their activity on the prostaglandin system. Altogether these findings ensure the rational use of BMY remedy in Thai traditional medicine.


Asunto(s)
Antiinflamatorios/farmacología , Nocicepción/efectos de los fármacos , Péptidos Opioides/efectos de los fármacos , Extractos Vegetales/farmacología , Raíces de Plantas , Prostaglandinas/metabolismo , Aegle , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Indometacina/farmacología , Masculino , Medicina Tradicional de Asia Oriental , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Naloxona/farmacología , Dimensión del Dolor , Preparaciones de Plantas/farmacología , Sapindaceae
4.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216445

RESUMEN

The role of TRPA1 receptor channels in meningeal nociception underlying the generation of headaches is still unclear. Activating as well as inhibitory effects of TRPA1 agonists have been reported in animal models of headache. The aim of the present study was to clarify the effect of the TRPA1 agonist nitroxyl (HNO) delivered by Angeli's salt in two rodent models of meningeal nociception. Single fibre recordings were performed using half-skull preparations of mice (C57BL/6) in vitro. Angeli's salt solution (AS, 300 µM) caused short-lasting vigorous increases in neuronal activity of primary meningeal afferents, followed by deactivation and desensitisation. These effects were similar in TRPA1 knockout and even more pronounced in TRPA1/TRPV1 double-knockout mice in comparison to wild-type mice. The activity of spinal trigeminal neurons with afferent input from the dura mater was recorded in vivo in anesthetised rats. AS (300 µM) or the TRPA1 agonist acrolein (100 and 300 µM) was applied to the exposed dura mater. AS caused no significant changes in spontaneous activity, while the mechanically evoked activity was reduced after acrolein application. These results do not confirm the assumption that activation of trigeminal TRPA1 receptor channels triggers the generation of headaches or contributes to its aggravation. Instead, there is evidence that TRPA1 activation may have an inhibitory function in the nociceptive trigeminal system.


Asunto(s)
Duramadre/efectos de los fármacos , Cefalea/tratamiento farmacológico , Neuronas Aferentes/efectos de los fármacos , Óxidos de Nitrógeno/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Duramadre/metabolismo , Femenino , Cefalea/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas Aferentes/metabolismo , Nocicepción/efectos de los fármacos , Ratas , Ratas Wistar , Canal Catiónico TRPA1/metabolismo , Canales Catiónicos TRPV/metabolismo , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo
5.
J Med Chem ; 65(3): 2522-2531, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34994550

RESUMEN

New psychoactive substance (NPS) opioids have proliferated within the international drug market. While synthetic opioids are traditionally composed of fentanyl analogues, benzimidazole-derived isotonitazene and its derivatives are the current NPS opioids of concern. Hence, in this study, we implement immunopharmacotherapy wherein antibodies are produced with high titers and nanomolar affinity to multiple benzimidazole-derived NPS opioids (BNO). Notably, these antibodies blunt psychoactive and physiological repercussions from BNO exposure, which was observed through antinociception, whole-body plethysmography, and blood-brain biodistribution studies. Moreover, we detail previously unreported pharmacokinetics of these drugs, which explains the struggle of traditional pharmaceutical opioid antagonists against BNO substances. These findings provide further insight into the in vivo effects of BNO drugs and the development of effective broad-spectrum therapeutics against NPS opioids.


Asunto(s)
Analgésicos Opioides/inmunología , Bencimidazoles/inmunología , Drogas Ilícitas/inmunología , Vacunas Conjugadas/inmunología , Analgésicos Opioides/síntesis química , Analgésicos Opioides/farmacocinética , Animales , Bencimidazoles/síntesis química , Bencimidazoles/farmacocinética , Femenino , Haptenos/química , Haptenos/inmunología , Hemocianinas/química , Hemocianinas/inmunología , Drogas Ilícitas/síntesis química , Drogas Ilícitas/farmacocinética , Ratones Endogámicos BALB C , Nocicepción/efectos de los fármacos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/prevención & control , Vacunas Conjugadas/química
6.
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
7.
Behav Brain Res ; 416: 113565, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34499933

RESUMEN

Acid-sensing ion channels (ASICs) play significant roles in numerous neurological and pathological conditions, including pain. Although acid-induced nociception has been characterized previously in zebrafish, the contribution of ASICs in modulating pain-like behaviors is still unknown. Here, we investigated the role of amiloride, a nonselective ASICs blocker, in the negative modulation of specific behavioral responses in a zebrafish-based model of acute visceral pain. We verified that intraperitoneal injection (i.p.) of 0.25, 0.5, 1.0, and 2.0 mg/mL amiloride alone or vehicle did not change zebrafish behavior compared to saline-treated fish. Administration of 2.5% acetic acid (i.p.) elicited writhing-like response evidenced by the abnormal body curvature and impaired locomotion and motor activity. Attenuation of acetic acid-induced pain was verified at lower amiloride doses (0.25 and 0.5 mg/mL) whereas 1.0 and 2.0 mg/mL abolished pain-like responses. The protective effect of the highest amiloride dose tested was evident in preventing writhing-like responses and impaired locomotion and vertical activity. Collectively, amiloride antagonized abdominal writhing-like phenotype and aberrant behaviors, supporting the involvement of ASICs in a zebrafish-based model of acute visceral pain.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Amilorida/farmacología , Locomoción/efectos de los fármacos , Dolor Visceral/tratamiento farmacológico , Pez Cebra , Animales , Modelos Animales de Enfermedad , Inyecciones Intraperitoneales , Masculino , Nocicepción/efectos de los fármacos
8.
Behav Brain Res ; 417: 113595, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34592375

RESUMEN

It has been shown that kappa opioid receptor (KOR) antagonists, such as nor-binaltorphimine (nor-BNI), have antinociceptive effects in some pain models that affect the trigeminal system. Also, its anxiolytic-like effect has been extensively demonstrated in the literature. The present study aimed to investigate the systemic, local, and central effect of nor-BNI on trigeminal neuropathic pain using the infraorbital nerve constriction model (CCI-ION), as well as to evaluate its effect on anxiety-like behavior associated with this model. Animals received nor-BNI systemically; in the trigeminal ganglion (TG); in the subarachnoid space to target the spinal trigeminal nucleus caudalis (Sp5C) or in the central amygdala (CeA) 14 days after CCI-ION surgery. Systemic administration of nor-BNI caused a significant reduction of facial mechanical hyperalgesia and promoted an anxiolytic-like effect, which was detected in the elevated plus-maze and the light-dark transition tests. When administered in the TG or CeA, the KOR antagonist was able to reduce facial mechanical hyperalgesia induced by CCI-ION, but without changing the anxiety-like behavior. Moreover, no change was observed on nociception and anxiety-like behavior after nor-BNI injection into the Sp5C. The present study demonstrated antinociceptive and anxiolytic-like effects of nor-BNI in a model of trigeminal neuropathic pain. The antinociceptive effect seems to be dissociated from the anxiolytic-like effect, at both the sites involved and at the dose need to achieve the effect. In conclusion, the kappa opioid system may represent a promising target to be explored for the control of trigeminal pain and associated anxiety. However, further studies are necessary to better elucidate its functioning and modulatory role in chronic trigeminal pain states.


Asunto(s)
Ansiedad/tratamiento farmacológico , Dolor Crónico/complicaciones , Hiperalgesia/tratamiento farmacológico , Naltrexona/análogos & derivados , Receptores Opioides kappa/antagonistas & inhibidores , Neuralgia del Trigémino/complicaciones , Animales , Núcleo Amigdalino Central/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Naltrexona/farmacología , Nocicepción/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
9.
Neuropharmacology ; 205: 108925, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921830

RESUMEN

The abuse of oral formulations of prescription opioids has precipitated the current opioid epidemic. We developed an oral oxycodone consumption model consisting of a limited access (4 h) two-bottle choice drinking in the dark (TBC-DID) paradigm and quantified dependence with naloxone challenge using mice of both sexes. We also assessed neurobiological correlates of withdrawal and dependence elicited via oral oxycodone consumption using immunohistochemistry for DeltaFosB (ΔFosB), a transcription factor described as a molecular marker for drug addiction. Neither sex developed a preference for the oxycodone bottle, irrespective of oxycodone concentration, bottle position or prior water restriction. Mice that volitionally consumed oxycodone exhibited hyperlocomotion in an open field test and supraspinal but not spinally-mediated antinociception. Both sexes also developed robust, dose-dependent levels of opioid withdrawal that was precipitated by the opioid antagonist naloxone. Oral oxycodone consumption followed by naloxone challenge led to mesocorticolimbic region-dependent increases in the number of ΔFosB expressing cells. Naloxone-precipitated withdrawal jumps, but not the oxycodone bottle % preference, was positively correlated with the number of ΔFosB expressing cells specifically in the nucleus accumbens shell. Thus, limited access oral consumption of oxycodone produced physical dependence and increased ΔFosB expression despite the absence of opioid preference. Our TBC-DID paradigm allows for the study of oral opioid consumption in a simple, high-throughput manner and elucidates the underlying neurobiological substrates that accompany opioid-induced physical dependence.


Asunto(s)
Analgésicos Opioides/farmacología , Núcleo Accumbens/efectos de los fármacos , Trastornos Relacionados con Opioides , Oxicodona/farmacología , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Síndrome de Abstinencia a Sustancias , Analgésicos Opioides/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Locomoción/efectos de los fármacos , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Nocicepción/efectos de los fármacos , Núcleo Accumbens/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/fisiopatología , Oxicodona/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología
10.
Behav Brain Res ; 420: 113728, 2022 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-34952028

RESUMEN

Traditional and novel analgesic modalities have been extensively tested for post-craniotomy pain management, yet the role of newer antiepileptic drugs in this area remains obscure. This study investigates the impact of levetiracetam (LEV) on pain modulation and neurobehavioral performance in a craniotomy model. Fifty-six Wistar rats were randomly assigned into seven groups: no intervention (CTRL), administration of placebo or LEV with no further intervention (PBO and LEV, respectively), and sham-operation or craniotomy in placebo (PBO-SHAM and PBO-CR, respectively) or LEV-treated rats (LEV-SHAM and LEV-CR, respectively). Pain was assessed by the rat grimace scale before, and at 8 and 24 h after craniotomy, following intraperitoneal injections of LEV (100 mg/kg twice daily) or normal saline two consecutive days before and on the craniotomy day. Elevated plus-maze and olfactory social memory tests were performed at 24- and 48 h post-craniotomy, respectively. Upon testing conclusion blood samples were collected for cytokines estimation. Levetiracetam administration enhanced antinociception in sham and craniotomy groups. In the elevated plus-maze test, LEV-CR rats spent more time in investigating open arms and performed more open arm entries than PBO-SHAM and PBO-CR animals. The olfactory test revealed no between-groups difference in acquisition time during first contact with a juvenile rat, while LEV-CR rats spent less time to recognize the same juvenile rat compared to PBO-SHAM and PBO-CR groups. Furthermore, LEV-treatment attenuated cortisol, interleukin-6 and TNF-a release, in sham and craniotomy animals. In conclusion, preemptive use of LEV decreases nociception, improves pain-evoked behavior and attenuates stress response in rats subjected to craniotomy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Craneotomía/efectos adversos , Levetiracetam/uso terapéutico , Dolor/tratamiento farmacológico , Animales , Masculino , Nocicepción/efectos de los fármacos , Ratas , Ratas Wistar
11.
Neuropeptides ; 91: 102212, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34826712

RESUMEN

Orofacial pain is one of the most common medical challenges. A preliminary report indicates that the NOP receptor may act as a therapeutic target in orofacial pain. Previous studies have shown that [(pF)Phe4, Aib7, Aib11, Arg14, Lys15]N/OFQ-NH2 (NOP01) functions as a potent NOP receptor peptide agonist. This work aims to investigate the antinociception of NOP01 and its possible action mechanisms in a formalin-induced mouse orofacial pain model at different levels. Our results demonstrated that local, intraperitoneal (i.p.) or intrathecal (i.t.) injection of NOP01 produced dose-related antinociception in both phases of the formalin pain, which could be inhibited by the NOP receptor antagonist but not the classical opioid receptor antagonist. Furthermore, the antinociception induced by systemic NOP01 was blocked by local but not spinal pretreatment with the NOP receptor antagonist, suggesting the involvement of the peripheral NOP receptor in NOP01-induced systemic antinociception. Moreover, local injection of NOP01 markedly suppressed the expression of c-Fos protein induced by formalin in ipsilateral trigeminal ganglion (TG) neurons. In conclusion, this work suggests that NOP01 exerts significant antinociception on orofacial pain at both peripheral and spinal levels via the NOP receptor. Notably, NOP01 cannot readily penetrate the blood-brain barrier. Thus, NOP01 may behave as a potential compound for developing peripherally restricted analgesics.


Asunto(s)
Analgésicos/uso terapéutico , Dolor Facial/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Receptores Opioides/agonistas , Analgésicos/farmacología , Animales , Modelos Animales de Enfermedad , Dolor Facial/inducido químicamente , Formaldehído , Ratones , Dimensión del Dolor , Receptor de Nociceptina
12.
CNS Neurosci Ther ; 28(2): 289-297, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34862748

RESUMEN

AIMS: The α2 -adrenergic receptor (α2 -AR) agonists have been shown to be effective in the treatment of various pain. For example, dexmedetomidine (DEX), a selective α2A -AR agonist, can be used for peripheral analgesia. However, it is not yet fully elucidated for the precise molecular mechanisms. P2X3 receptor is a major receptor processing nociceptive information in primary sensory neurons. Herein, we show that a functional interaction of α2A -ARs and P2X3 receptors in dorsal root ganglia (DRG) neurons could contribute to peripheral analgesia of DEX. METHODS: Electrophysiological recordings were carried out on rat DRG neurons, and nociceptive behavior was quantified in rats. RESULTS: The activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated and α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents in a concentration-dependent and voltage-independent manner. Pre-application of DEX shifted the α,ß-meATP concentration-response curve downwards, with a decrease of 50.43 ± 4.75% in the maximal current response of P2X3 receptors to α,ß-meATP in the presence of DEX. Suppression of α,ß-meATP-evoked currents by DEX was blocked by the α2A -AR antagonist BRL44408 and prevented by intracellular application of the Gi/o protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. DEX also suppressed α,ß-meATP-evoked action potentials through α2A -ARs in rat DRG neurons. Finally, the activation of peripheral α2A -ARs by DEX had an analgesic effect on the α,ß-meATP-induced nociception. CONCLUSIONS: These results suggested that activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated electrophysiological and behavioral activity via a Gi/o proteins and cAMP signaling pathway, which was a novel potential mechanism underlying analgesia of peripheral α2A -AR agonists.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Nocicepción/efectos de los fármacos , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Receptores Purinérgicos P2X3/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Dexmedetomidina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley
13.
Neuropharmacology ; 205: 108924, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34919904

RESUMEN

Peripheral A1 adenosine receptor signaling has been shown to have analgesic effects in a variety of pain conditions. However, it is not yet fully elucidated for the precise molecular mechanisms. Acid sensing ion channels (ASICs) are expressed predominantly in nociceptive sensory neurons responding to protons. Given that both A1 adenosine receptors and ASICs are present in dorsal root ganglia (DRG) neurons, we therefore investigated whether there was a cross-talk between the two types of receptors. Herein, electrophysiological recordings showed that the A1 adenosine receptor agonist N6-cyclopentyladenosine (CPA) suppressed acid-induced currents and action potentials, which were mediated by ASICs, in rat DRG neurons. CPA inhibited the maximum response to protons, as shown a downward shift of concentration-response curve for protons. The CPA-induced suppression of ASIC currents was blocked by the A1 adenosine receptor antagonist KW-3902 and also prevented by intracellular application of the Gi/o-protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. Finally, intraplantar pretreatment of CPA dose-dependently relieved acid-induced nociceptive responses in rats through peripheral A1 adenosine receptors. These results suggested that CPA suppressed ASICs via A1 adenosine receptors and intracellular Gi/o-proteins and cAMP signaling cascades in rat DRG neurons, which was a novel potential mechanism underlying analgesia of peripheral A1 adenosine receptors.


Asunto(s)
Canales Iónicos Sensibles al Ácido/efectos de los fármacos , Agonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A1/farmacología , Analgesia , Fenómenos Electrofisiológicos/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Nocicepción/efectos de los fármacos , Nociceptores/efectos de los fármacos , Receptor de Adenosina A1/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Ratas
14.
Neuropharmacology ; 202: 108858, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34715121

RESUMEN

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Asunto(s)
Tolerancia a Medicamentos/genética , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/fisiología , Dependencia de Morfina/genética , Morfina/efectos adversos , Nocicepción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/genética , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/terapia
15.
Sci Rep ; 11(1): 24358, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34934106

RESUMEN

The present study has explored the hypothesis that neurokinin1 receptors (NK1Rs) in medial septum (MS) modulate nociception evoked on hind paw injection of formalin. Indeed, the NK1Rs in MS are localized on cholinergic neurons which have been implicated in nociception. In anaesthetized rat, microinjection of L-733,060, an antagonist at NK1Rs, into MS antagonized the suppression of CA1 population spike (PS) evoked on peripheral injection of formalin or on intraseptal microinjection of substance P (SP), an agonist at NK1Rs. The CA1 PS reflects the synaptic excitability of pyramidal cells in the region. Furthermore, microinjection of L-733,060 into MS, but not LS, attenuated formalin-induced theta activation in both anaesthetized and awake rat, where theta reflects an oscillatory information processing by hippocampal neurons. The effects of L-733,060 on microinjection into MS were nociceptive selective as the antagonist did not block septo-hippocampal response to direct MS stimulation by the cholinergic receptor agonist, carbachol, in anaesthetized animal or on exploration in awake animal. Interestingly, microinjection of L-733,060 into both MS and LS attenuated formalin-induced nociceptive flinches. Collectively, the foregoing novel findings highlight that transmission at NK1R provide an affective valence to septo-hippocampal information processing and that peptidergic transmission in the septum modulates nociceptive behaviours.


Asunto(s)
Formaldehído/toxicidad , Inflamación/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Piperidinas/farmacología , Prosencéfalo/efectos de los fármacos , Receptores de Neuroquinina-1/química , Núcleos Septales/efectos de los fármacos , Animales , Desinfectantes/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Dolor/inducido químicamente , Dolor/metabolismo , Dolor/patología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo , Núcleos Septales/metabolismo , Núcleos Septales/patología
16.
PLoS One ; 16(12): e0260423, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34879087

RESUMEN

Complete Freund's adjuvant (CFA) has been used to develop the arthritic or inflammatory condition in the animal, but there is a lack of information concerning high CFA doses on nociceptive behaviour and inflammatory parameters. This study aimed to compare the effects of different high doses of CFA in rat to closely mimic nociceptive and inflammatory parameters of rheumatoid arthritis (RA) in humans. Twenty-four male Sprague-Dawley rats were randomly divided into four groups (n = 6): Control (C), CFA-induced polyarthritic groups at 5.0 mg/mL (CFA 5.0), 7.5 mg/mL (CFA 7.5) and 10.0mg/mL (CFA 10.0). The rats' right hindpaw was inoculated with CFA intradermally and developed into a polyarthritic state within 20 days. Nociceptive behavioural assessments, including von Frey and hot plate tests and spontaneous activities, were conducted on day 0, 7, 15 and 20. Bilateral ankle joints diameter and circumference, full blood count, joints and paw histological examinations were also conducted throughout the study period. Based on the results, CFA 5.0 and CFA 7.5 groups showed a significant increase in spontaneous activities and development of thermal hyperalgesia but no change in body weight and food intake, no development of tactile allodynia and haematological indices, and no significant morphological changes of joints histology. Meanwhile, CFA 10.0 group demonstrated significant and constant changes in all nociceptive and inflammatory parameters investigated. In conclusion, CFA at the dose of 10mg/mL has the most potential and reliable dosage to develop polyarthritis in a rat model to mimic RA condition in humans.


Asunto(s)
Artritis Experimental/fisiopatología , Artritis Reumatoide/fisiopatología , Adyuvante de Freund/efectos adversos , Nocicepción/efectos de los fármacos , Animales , Artritis Experimental/inducido químicamente , Artritis Reumatoide/inducido químicamente , Relación Dosis-Respuesta a Droga , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
17.
J Neuroinflammation ; 18(1): 279, 2021 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857006

RESUMEN

BACKGROUND: Opioid analgesics remain widely used for pain treatment despite the related serious side effects. Some of those, such as opioid tolerance and opioid-induced hyperalgesia may be at least partially due to modulation of opioid receptors (OR) function at nociceptive synapses in the spinal cord dorsal horn. It was suggested that increased release of different chemokines under pathological conditions may play a role in this process. The goal of this study was to investigate the crosstalk between the µOR, transient receptor potential vanilloid 1 (TRPV1) receptor and C-C motif ligand 2 (CCL2) chemokine and the involvement of spinal microglia in the modulation of opioid analgesia. METHODS: Patch-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and dorsal root evoked currents (eEPSC) in spinal cord slices superficial dorsal horn neurons were used to evaluate the effect of µOR agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), CCL2, TRPV1 antagonist SB366791 and minocycline. Paw withdrawal test to thermal stimuli was combined with intrathecal (i.t.) delivery of CCL2 and DAMGO to investigate the modulation in vivo. RESULTS: Application of DAMGO induced a rapid decrease of mEPSC frequency and eEPSC amplitude, followed by a delayed increase of the eESPC amplitude, which was prevented by SB366791. Chemokine CCL2 treatment significantly diminished all the DAMGO-induced changes. Minocycline treatment prevented the CCL2 effects on the DAMGO-induced eEPSC depression, while mEPSC changes were unaffected. In behavioral experiments, i.t. injection of CCL2 completely blocked DAMGO-induced thermal hypoalgesia and intraperitoneal pre-treatment with minocycline prevented the CCL2 effect. CONCLUSIONS: Our results indicate that opioid-induced inhibition of the excitatory synaptic transmission could be severely attenuated by increased CCL2 levels most likely through a microglia activation-dependent mechanism. Delayed potentiation of neurotransmission after µOR activation is dependent on TRPV1 receptors activation. Targeting CCL2 and its receptors and TRPV1 receptors in combination with opioid therapy could significantly improve the analgesic properties of opioids, especially during pathological states.


Asunto(s)
Analgésicos Opioides/farmacología , Quimiocina CCL2/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Nocicepción/efectos de los fármacos , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Anilidas/farmacología , Animales , Cinamatos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar
18.
Biol Pharm Bull ; 44(11): 1746-1751, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34719650

RESUMEN

Tramadol is a weak opioid that produces analgesic effect via both the µ-opioid receptor (MOR) and non-opioid targets. Constipation is the most common opioid-related side effect in patients with cancer and non-cancer pain. However, the contribution of MOR to tramadol-induced constipation is unclear. Therefore, we used naldemedine, a peripherally acting MOR antagonist, and MOR-knockout mice to investigate the involvement of peripheral MOR in tramadol-induced constipation using a small intestinal transit model. A single dose of tramadol (3-100 mg/kg, per os (p.o.)) inhibited small intestinal transit dose-dependently in rats. Naldemedine (0.01-10 mg/kg, p.o.) blocked the inhibition of small intestinal transit induced by tramadol (30 mg/kg, p.o.) in rats. The transition rate increased dose-dependently over the range of naldemedine 0.01-0.3 mg/kg, and complete recovery was observed at 0.3-10 m/kg. Additionally, tramadol (30 and 100 mg/kg, subcutaneously (s.c.)) inhibited small intestinal transit in wild-type mice but not in MOR-knockout mice. These results suggest that peripheral MOR participates in tramadol-induced constipation.


Asunto(s)
Analgésicos Opioides/efectos adversos , Estreñimiento Inducido por Opioides/etiología , Receptores Opioides mu/efectos de los fármacos , Tramadol/efectos adversos , Analgésicos Opioides/sangre , Analgésicos Opioides/farmacocinética , Animales , Intestino Delgado/efectos de los fármacos , Masculino , Naltrexona/efectos adversos , Naltrexona/análogos & derivados , Naltrexona/sangre , Naltrexona/farmacocinética , Nocicepción/efectos de los fármacos , Estreñimiento Inducido por Opioides/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores Opioides mu/metabolismo , Tramadol/sangre , Tramadol/farmacocinética
19.
Neurotox Res ; 39(6): 1782-1799, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34792764

RESUMEN

Oxaliplatin-induced neurotoxicity is expressed as a dose-limiting peripheral sensory neuropathy (PSN). Cannabinoid substances have been investigated for the analgesic effect. This study aimed to investigate the role of cannabinoid receptors in oxaliplatin-associated PSN. Swiss male mice received nine oxaliplatin injections (2 mg/kg, i.v.). Mechanical and thermal nociceptive tests were performed for 56 days. CB1, CB2, and c-Fos expression were assessed in dorsal root ganglia (DRG), spinal cord (SC), trigeminal ganglia (TG), spinal trigeminal nucleus caudalis (Sp5C), and periaqueductal gray (PAG). Iba-1 expression was assessed in DRG and ATF3 in TG. Cannabidiol (10 mg/kg, p.o.) or a CB1/CB2 non-selective agonist (WIN 55,212-2; 0.5 mg/kg, s.c.) or AM251 (CB1 antagonist) or AM630 (CB2 antagonist) (3 mg/kg, i.p.) were injected before oxaliplatin. Oxaliplatin increased CB1 in DRG, SC, TG, Sp5C, and ventrolateral PAG, with no interference in CB2 expression. Cannabidiol increased CB1 in DRG, reduced mechanical hyperalgesia and c-Fos expression in DRG and SC. Additionally, WIN 55,212-2 increased CB1 in DRG, reduced mechanical hyperalgesia, cold allodynia and c-Fos expression in DRG and SC. CB1 blockage hastened the cold allodynia response, but the CB2 antagonist failed to modulate the oxaliplatin-induced nociceptive behavior. Oxaliplatin also increased Iba-1 in DRG, suggesting immune response modulation which was reduced by cannabidiol and enhanced by AM630. The modulation of the endocannabinoid system, through the CB1 receptor, attenuates the oxaliplatin-associated PNS. The activation of the endocannabinoid system could be considered as a therapeutic target for controlling oxaliplatin-associated neuropathy.


Asunto(s)
Endocannabinoides/metabolismo , Nocicepción/efectos de los fármacos , Oxaliplatino/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Receptor Cannabinoide CB1/agonistas , Animales , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Masculino , Ratones , Oxaliplatino/antagonistas & inhibidores , Dimensión del Dolor , Enfermedades del Sistema Nervioso Periférico/metabolismo , Receptor Cannabinoide CB1/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante
20.
Life Sci ; 285: 120014, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34619167

RESUMEN

AIMS: We have shown that chemokines injected into the periaqueductal gray region of the brain blocks opioid-induced analgesia in the rat cold-water tail flick test (CWTF). The present experiments tested whether chemokine receptor antagonists (CRAs), in combination with sub-analgesic doses of morphine, would provide maximal analgesia in the CWTF test and the mouse formalin pain assay. The effect of CRAs on respiratory depression was also evaluated. MAIN METHODS: One, two or four CRAs (AMD3100/CXCR4, maraviroc/CCR5, RS504393/CCR2 orAZD8797/CX3CR1) were used in combination with sub-analgesic doses of morphine, all given systemically. Pain was assessed using the rat CWTF test or formalin injection into the paw of mice scored by licking. Respiration and oxygen saturation were measured in rats using a MouseOX® Plus - pulse oximeter. KEY FINDINGS: In the CWTF test, a sub-maximal dose of morphine in combination with maraviroc alone, maraviroc plus AMD3100, or with the four chemokine receptor antagonists, produced synergistic increases in antinociception. In the formalin test, the combination of four CRAs plus a sub-maximal dose of morphine resulted in increased antinociception in both male and female mice. AMD3100 had an additive effect with morphine in both sexes. Coadministration of CRAs with morphine did not potentiate the opioid respiratory depressive effect. SIGNIFICANCE: These results support the conclusion that combinations of CRAs can increase the potency of sub-analgesic doses of morphine analgesia without increasing respiratory depression. The results support an "opioid sparing" strategy for alleviation of pain using reduced doses of opioids in combination with CRAs to achieve maximal analgesia.


Asunto(s)
Analgesia/métodos , Analgésicos Opioides/farmacología , Morfina/farmacología , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/tratamiento farmacológico , Receptores de Quimiocina/antagonistas & inhibidores , Animales , Bencilaminas/administración & dosificación , Bencilaminas/farmacología , Ciclamas/administración & dosificación , Ciclamas/farmacología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Masculino , Maraviroc/administración & dosificación , Maraviroc/farmacología , Morfina/administración & dosificación , Morfina/efectos adversos , Dolor Nociceptivo/fisiopatología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Insuficiencia Respiratoria/inducido químicamente , Tiazoles/administración & dosificación , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA