Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Life Sci ; 355: 123002, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39173999

ABSTRACT

It is known that the cannabinoid type 2 (CB2) receptor has an anti-inflammatory role. Therefore, animals without CB2 receptors show enhanced inflammation and pain in the model of chronic pain, e.g., neuropathic pain. We previously proposed the upregulated leptin signaling at the peripheral nerve as one of the underlying molecular mechanisms of pain exacerbation in nerve-injured CB2 knockouts, as they displayed robust upregulation of leptin receptors and leptin signaling in the peripheral nerve. Due to these past results, we hypothesized that CB2 receptor deficiency might also modify the peripheral neuroinflammation led by chronic exposure to a high-fat diet (HFD). Interestingly, CB2 knockout animals showed significant resistance to HFD-induced neuroinflammation. Namely, 5-week feeding of HFD induced substantial hypersensitivity in WT animals, while tactile sensitivity of HFD-fed CB2 knockouts remained intact. HFD-fed WT animals also displayed the robust upregulation of chemokine CXCR4 expression with increased macrophage infiltration, which was never observed in HFD-fed CB2 knockout mice. Moreover, 5-week HFD exposure led significant increase of CD11b+Ly6G-Ly6Chigh cells and a decrease of CD11b+Ly6G+Ly6Clow cells in the spleen of WT animals, which was also not found in either HFD-fed CB2 knockouts or standard diet-fed WT and CB2 animals. Together with past reports, these results suggest that CB2 receptors might have a double-sided regulatory role in the context of inflammation development or, more widely, immune system regulation. We propose that CB2 signaling is not always anti-inflammatory and could take a pro-inflammatory role depending on the cause of the inflammation.


Subject(s)
Diet, High-Fat , Mice, Inbred C57BL , Mice, Knockout , Neuroinflammatory Diseases , Receptor, Cannabinoid, CB2 , Animals , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Diet, High-Fat/adverse effects , Mice , Male , Neuroinflammatory Diseases/metabolism , Inflammation/metabolism
2.
Brain Res Bull ; 160: 40-49, 2020 07.
Article in English | MEDLINE | ID: mdl-32294520

ABSTRACT

Endocannabinoid system activity contributes to the homeostatic defense against aging and thus may counteract the progression of brain aging. The cannabinoid type 1 (CB1) receptor activity declines with aging in the brain, which impairs neuronal network integrity and cognitive functions. However, the underlying mechanisms that link CB1 activity and memory decline remain unknown. Mitochondrial activity profoundly influences neuronal function, and age-dependent mitochondrial activity change is one of the known hallmarks of brain aging. As CB1 receptor is expressed on mitochondria and may regulate neuronal energy metabolism in hippocampus, we hypothesized that CB1 receptors might influence mitochondria in hippocampal neurons. Here, we found that CB1 receptor significantly affected mitochondrial autophagy (mitophagy) and morphology in an age-dependent manner. Serine 65-phosphorylated ubiquitin, a key marker for mitophagy, was reduced in adult CB1-deficient mice (CB1-KO) compared to those in wild type controls, particularly in CA1 pyramidal cell layer. Transmission electron microscopy (TEM) analysis showed reduced mitophagy-like events in hippocampus of adult CB1-KO. TEM analysis also showed that mitochondrial morphology in adult CB1-KO mice was altered shown by an increase in thin and elongated mitochondria in hippocampal neurons. 3D reconstruction of mitochondrial morphology after scanning electron microscopy additionally revealed an enhanced density of interconnected mitochondria. Altogether, these findings suggest that reduced CB1 signaling in CB1-KO mice leads to reduced mitophagy and abnormal mitochondrial morphology in hippocampal neurons during aging. These mitochondrial changes might be due to the impairments in mitochondrial quality control system, which links age-related decline in CB1 activity and impaired memory.


Subject(s)
Aging/metabolism , Autophagy/physiology , Hippocampus/metabolism , Mitochondrial Dynamics/physiology , Neurons/metabolism , Receptor, Cannabinoid, CB1/deficiency , Aging/pathology , Animals , Female , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology
3.
Clin Lab ; 66(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-32013358

ABSTRACT

BACKGROUND: Preterm infants sometimes have transient late-onset hemolytic jaundice; however, the etiology has yet to be determined. CASE PRESENTATION: In our case, fetal hemoglobin (HbF) level increased significantly to 100% at 23 days of age. Levels of methemoglobin and carboxyhemoglobin also increased to 2.9% and 3.5%, respectively, following the elevated HbF level. At 26 days, hemolytic jaundice developed. No abnormality of red blood cell membranes and enzyme activities was found. CONCLUSIONS: The etiology of late-onset hemolytic jaundice in preterm infants may associate with an impaired switching from HbF to adult hemoglobin (HbA) or reverse switching from HbA to HbF.


Subject(s)
Fetal Hemoglobin/analysis , Infant, Extremely Low Birth Weight , Jaundice , Erythrocytes/pathology , Humans , Infant, Newborn , Male , Methemoglobin/analysis
4.
Sci Rep ; 9(1): 7468, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097758

ABSTRACT

Neuropathic pain can develop after nerve injury, leading to a chronic condition with spontaneous pain and hyperalgesia. Pain is typically restricted to the side of the injured nerve, but may occasionally spread to the contralateral side, a condition that is often referred to as mirror-image pain. Mechanisms leading to mirror-image pain are not completely understood, but cannabinoid CB2 receptors have been implicated. In this study, we use genetic mouse models to address the question if CB2 receptors on neurons or on microglia/macrophages are involved. First, we show that a GFP reporter protein under control of the CB2 promoter is induced upon partial sciatic nerve ligation in spinal cord, dorsal root ganglia, and highest in sciatic nerve macrophages, but not in neurons. Mice which lack CB2 receptors specifically on myeloid cells (microglia, macrophages) developed a mirror-image allodynia [treatment F1,48 = 45.69, p < 0.0001] similar to constitutive CB2 receptor knockout mice [treatment F1,70 = 92.41, p < 0.0001]. Such a phenotype was not observed after the deletion of CB2 from neurons [treatment F1,70 = 0.1315, p = 0.7180]. This behavioral pain phenotype was accompanied by an increased staining of microglia in the dorsal horn of the spinal cord, as evidenced by an enhanced Iba 1 expression [CB2KO, p = 0.0175; CB2-LysM, p = 0.0425]. Similarly, myeloid-selective knockouts showed an increased expression of the leptin receptor in the injured ipsilateral sciatic nerve, thus further supporting the notion that leptin signaling contributes to the increased neuropathic pain responses of CB2 receptor knockout mice. We conclude that CB2 receptors on microglia and macrophages, but not on neurons, modulate neuropathic pain responses.


Subject(s)
Hyperalgesia/metabolism , Macrophages/metabolism , Neuralgia/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Calcium-Binding Proteins/metabolism , Gene Deletion , Hyperalgesia/physiopathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/physiology , Neuralgia/physiopathology , Neurons/metabolism , Neurons/physiology , Receptor, Cannabinoid, CB2/genetics , Receptors, Leptin/metabolism
5.
Sci Rep ; 8(1): 10827, 2018 Jul 17.
Article in English | MEDLINE | ID: mdl-30018366

ABSTRACT

Neuropathic pain typically appears in a region innervated by an injured or diseased nerve and, in some instances, also on the contralateral side. This so-called mirror image pain is often observed in mice lacking CB2 receptors after sciatic nerve injury, but the underlying mechanisms for this phenotype largely remain unclear. Here we focused on peripheral leptin signaling, which modulates neuropathic pain development and interacts with the endocannabinoid system. Leptin production is induced at the site of nerve injury in CB2-deficient mice (CB2-KO) mice and wild type controls (WT). However, induction of leptin receptor expression was only observed in the injured nerve of CB2-KO mice. This was paralleled by a stimulation of the leptin receptor-downstream STAT3 signaling and an infiltration of F4/80-positive macrophages. Interestingly, an upregulation of leptin receptor expression STAT3 activity and macrophage infiltration was also observed on the non-injured nerve of CB2-KO mice thus reflecting the mirror image pain in CB2-KO animals. Importantly, perineurally-administered leptin-neutralizing antibodies reduced mechanical hyperalgesia, blocked mirror image pain and inhibited the recruitment of F4/80-positive macrophages. These results identify peripheral leptin signaling as an important modulator of CB2 signaling in neuropathic pain.


Subject(s)
Leptin/metabolism , Neuralgia/pathology , Receptor, Cannabinoid, CB2/metabolism , Animals , Antibodies, Neutralizing/administration & dosage , Hyperalgesia/drug therapy , Leptin/immunology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/drug therapy , Neuralgia/etiology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/pathology , Receptor, Cannabinoid, CB2/genetics , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Sciatic Nerve/metabolism , Signal Transduction , Up-Regulation
6.
Eur Neuropsychopharmacol ; 25(8): 1388-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25910421

ABSTRACT

There is evidence to suggest that a dysregulation of endocannabinoid signaling may contribute to the etiology and pathophysiology of migraine. Thus, patients suffering from chronic migraine or medication overuse headache showed alterations in the activity of the arachidonoylethanolamide (AEA) degrading enzyme fatty acid amide hydrolase (FAAH) and a specific AEA membrane transporter, alongside with changes in AEA levels. The precise role of different endocannabinoid system components is, however, not clear. We have therefore investigated mice with a genetic deletion of the two main cannabinoid receptors CB1 and CB2, or the main endocannabinoid degrading enzymes, FAAH and monoacylglycerol lipase (MAGL), which degrades 2-arachidonoylglycerol (2-AG), in a nitroglycerine-induced animal model of migraine. We found that nitroglycerin-induced mechanical allodynia and neuronal activation of the trigeminal nucleus were completely abolished in FAAH-deficient mice. To validate these results, we used two structurally different FAAH inhibitors, URB597 and PF3945. Both inhibitors also dose-dependently blocked nitroglycerin-induced hyperalgesia and the activation of trigeminal neurons. The effects of the genetic deletion of pharmacological blockade of FAAH are mediated by CB1 receptors, because they were completely disrupted with the CB1 antagonist rimonabant. These results identify FAAH as a target for migraine pharmacotherapy.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Hyperalgesia/drug therapy , Migraine Disorders/drug therapy , Trigeminal Nuclei/drug effects , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Arachidonic Acids , Benzamides/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endocannabinoids , Enzyme Inhibitors/pharmacology , Hyperalgesia/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Migraine Disorders/physiopathology , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Nitroglycerin , Pain Measurement , Piperidines/pharmacology , Polyunsaturated Alkamides , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Rimonabant , Touch , Trigeminal Nuclei/physiopathology
7.
J Pharmacol Exp Ther ; 342(3): 799-807, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22700431

ABSTRACT

N,N-diethyl-4-(5-hydroxyspiro[chromene-2,4'-piperidine]-4-yl) benzamide (ADL5859) and N,N-diethyl-3-hydroxy-4-(spiro[chromene-2,4'-piperidine]-4-yl)benzamide (ADL5747) are novel δ-opioid agonists that show good oral bioavailability and analgesic and antidepressive effects in the rat and represent potential drugs for chronic pain treatment. Here, we used genetic approaches to investigate molecular mechanisms underlying their analgesic effects in the mouse. We tested analgesic effects of ADL5859 and ADL5747 in mice by using mechanical sensitivity measures in both complete Freund's adjuvant and sciatic nerve ligation pain models. We examined their analgesic effects in δ-opioid receptor constitutive knockout (KO) mice and mice with a conditional deletion of δ-receptor in peripheral voltage-gated sodium channel (Nav)1.8-expressing neurons (cKO mice). Both ADL5859 and ADL5747, and the prototypical δ agonist 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethyl-piperazin-1-yl]-(3-methoxyphenyl)methyl]-N,N-diethyl-benzamide (SNC80) as a control, significantly reduced inflammatory and neuropathic pain. The antiallodynic effects of all three δ-opioid agonists were abolished in constitutive δ-receptor KO mice and strongly diminished in δ-receptor cKO mice. We also measured two other well described effects of δ agonists, increase in locomotor activity and agonist-induced receptor internalization by using knock-in mice expressing enhanced green fluorescence protein-tagged δ receptors. In contrast to SNC80, ADL5859 and ADL5747 did not induce either hyperlocomotion or receptor internalization in vivo. In conclusion, both ADL5859 and ADL5747 showed efficient pain-reducing properties in the two models of chronic pain. Their effects were mediated by δ-opioid receptors, with a main contribution of receptors expressed on peripheral Nav1.8-positive neurons. The lack of in vivo receptor internalization and locomotor activation, typically induced by SNC80, suggests agonist-biased activity at the receptor for the two drugs.


Subject(s)
Benzamides/pharmacology , Benzopyrans/pharmacology , Locomotion/drug effects , Neuralgia/drug therapy , Receptors, Opioid, delta/metabolism , Spiro Compounds/pharmacology , Analgesia/methods , Analgesics, Opioid/agonists , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Locomotion/genetics , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neuralgia/genetics , Neuralgia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Pain Measurement/methods , Piperazines/pharmacology , Receptors, Opioid, delta/genetics
8.
Trends Pharmacol Sci ; 32(10): 581-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21925742

ABSTRACT

Compared to the better-known mu opioid receptor, delta opioid receptors have been relatively understudied. However, the development of highly selective delta opioid agonists and the availability of genetic mouse models have extended our knowledge of delta opioid receptors in vivo. Here we review recent developments in the characterization of delta opioid receptor biology and aspects of delta opioid receptor function that have potential for therapeutic targeting. Preclinical data have confirmed that delta opioid receptor activation reduces persistent pain and improves negative emotional states; clinical trials have been initiated to assess the effectiveness of delta opioid agonists in chronic pain and depression. Furthermore, a possible role for these receptors in neuroprotection is being investigated. The usefulness of targeting delta opioid receptors in drug abuse remains open and a role for these receptors in impulse control disorders is emerging. Finally, the recent demonstration of biased agonism at the delta opioid receptor in vivo opens novel perspectives towards targeting specific therapeutic effects through drug design.


Subject(s)
Brain Diseases/drug therapy , Receptors, Opioid, delta/agonists , Animals , Brain Diseases/metabolism , Clinical Trials as Topic , Drug Design , Drug Evaluation, Preclinical , Humans , Receptors, Opioid, delta/metabolism
9.
Nat Neurosci ; 14(8): 1017-22, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725314

ABSTRACT

Zinc is abundant in the central nervous system and regulates pain, but the underlying mechanisms are unknown. In vitro studies have shown that extracellular zinc modulates a plethora of signaling membrane proteins, including NMDA receptors containing the NR2A subunit, which display exquisite zinc sensitivity. We created NR2A-H128S knock-in mice to investigate whether Zn2+-NR2A interaction influences pain control. In these mice, high-affinity (nanomolar) zinc inhibition of NMDA currents was lost in the hippocampus and spinal cord. Knock-in mice showed hypersensitivity to radiant heat and capsaicin, and developed enhanced allodynia in inflammatory and neuropathic pain models. Furthermore, zinc-induced analgesia was completely abolished under both acute and chronic pain conditions. Our data establish that zinc is an endogenous modulator of excitatory neurotransmission in vivo and identify a new mechanism in pain processing that relies on NR2A NMDA receptors. The study also potentially provides a molecular basis for the pain-relieving effects of dietary zinc supplementation.


Subject(s)
Neurons/drug effects , Pain/drug therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Trace Elements/pharmacology , Acoustic Stimulation , Analysis of Variance , Animals , DNA Mutational Analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Hand Strength/physiology , Hippocampus/cytology , Histidine/genetics , In Vitro Techniques , Larva , Locomotion/drug effects , Locomotion/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pain/etiology , Pain/physiopathology , Pain Measurement , Pain Threshold/drug effects , Pain Threshold/physiology , Physical Stimulation , Protein Binding/drug effects , Reaction Time/drug effects , Reaction Time/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Reflex/drug effects , Rotarod Performance Test/methods , Serine/genetics , Smell/drug effects , Smell/genetics , Spinal Cord/cytology , Statistics, Nonparametric , Touch Perception/drug effects , Touch Perception/genetics , Trace Elements/therapeutic use , Xenopus , Zinc/pharmacology , Zinc/therapeutic use
10.
Pain ; 152(6): 1238-1248, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21295407

ABSTRACT

Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.


Subject(s)
Ganglia, Spinal/pathology , Nociceptors/physiology , Pain/genetics , Pain/pathology , Receptors, Opioid, delta/deficiency , Analgesics, Opioid/therapeutic use , Analysis of Variance , Animals , Benzamides/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Freund's Adjuvant/adverse effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Inflammation/chemically induced , Inflammation/complications , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/genetics , NAV1.8 Voltage-Gated Sodium Channel , Nociceptors/drug effects , Pain/etiology , Pain Measurement/methods , Piperazines/therapeutic use , Protein Binding/drug effects , Protein Binding/genetics , Sodium Channels/genetics , Sodium Channels/metabolism , Sulfur Isotopes/pharmacokinetics
11.
J Neurosci ; 30(49): 16459-68, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21147985

ABSTRACT

δ-Opioid receptors are G-protein-coupled receptors that regulate nociceptive and emotional responses. It has been well established that distinct agonists acting at the same G-protein-coupled receptor can engage different signaling or regulatory responses. This concept, known as biased agonism, has important biological and therapeutic implications. Ligand-biased responses are well described in cellular models, however, demonstrating the physiological relevance of biased agonism in vivo remains a major challenge. The aim of this study was to investigate the long-term consequences of ligand-biased trafficking of the δ-opioid receptor, at both the cellular and behavioral level. We used δ agonists with similar binding and analgesic properties, but high [SNC80 ((+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide)]- or low [ARM390 (N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide)]-internalization potencies. As we found previously, a single SNC80-but not ARM390-administration triggered acute desensitization of the analgesic response in mice. However, daily injections of either compound over 5 d produced full analgesic tolerance. SNC80-tolerant animals showed widespread receptor downregulation, and tolerance to analgesic, locomotor and anxiolytic effects of the agonist. Hence, internalization-dependent tolerance developed, as a result of generalized receptor degradation. In contrast, ARM390-tolerant mice showed intact receptor expression, but δ-opioid receptor coupling to Ca²+ channels was abolished in dorsal root ganglia. Concomitantly, tolerance developed for agonist-induced analgesia, but not locomotor or anxiolytic responses. Therefore, internalization-independent tolerance was produced by anatomically restricted adaptations leading to pain-specific tolerance. Hence, ligand-directed receptor trafficking of the δ-opioid receptor engages distinct adaptive responses, and this study reveals a novel aspect of biased agonism in vivo.


Subject(s)
Analgesics/pharmacology , Drug Tolerance/physiology , Ligands , Pain Threshold/physiology , Receptors, Opioid, delta/metabolism , Analgesics/therapeutic use , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Brain/ultrastructure , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/genetics , Disease Models, Animal , Drug Interactions , Drug Tolerance/genetics , Female , Freund's Adjuvant , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/complications , Locomotion/drug effects , Locomotion/genetics , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/drug therapy , Pain/etiology , Pain Threshold/drug effects , Patch-Clamp Techniques/methods , Piperazines/pharmacology , Piperazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Binding/drug effects , Protein Transport/genetics , Protein Transport/physiology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Sensory Receptor Cells/drug effects , Spinal Cord/ultrastructure , Statistics, Nonparametric , Sulfur Isotopes/metabolism , Time Factors
12.
Eur J Pharmacol ; 566(1-3): 181-4, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17459370

ABSTRACT

The role of cyclin-dependent kinase 5 (Cdk5) in the capsaicin-induced cough reflex was examined in mice. Pretreatment with inhaled roscovitine, a selective Cdk5 inhibitor, at concentrations of 0.3 to 3 mM inhibited the number of capsaicin-induced coughs in a concentration-dependent manner. Pretreatment with inhaled roscovitine, at a concentration of 3 mM also slightly but significantly inhibited the number of citric acid-induced coughs. The number of capsaicin-induced coughs was significantly reduced when C-fibers were desensitized by the pretreatment with capsaicin. The number of citric acid-induced coughs was slightly but significantly reduced in capsaicin-pretreated mice as compared with that in naive mice. Although the inhalation of roscovitine at a concentration of 3 mM significantly reduced the number of citric acid-induced coughs in naive mice to the level observed in capsaicin-pretreated mice, roscovitine had no effect on the number of citric acid-induced coughs in capsaicin-pretreated mice. These results suggest that Cdk5-dependent factors are involved in C-fiber-mediated cough signaling.


Subject(s)
Antitussive Agents/pharmacology , Cough/drug therapy , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Animals , Capsaicin , Citric Acid , Cough/chemically induced , Male , Mice , Mice, Inbred ICR , Roscovitine
13.
Eur J Pharmacol ; 560(2-3): 160-2, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17292346

ABSTRACT

The effect of naloxonazine, a selective mu(1)-opioid receptor antagonist, on oxycodone-induced antinociception was examined in streptozotocin-induced diabetic mice. Oxycodone (5 mg/kg, s.c.) induced significant antinociception in both non-diabetic and diabetic mice. This antinociceptive effect of oxycodone was completely antagonized by pretreatment with naloxonazine (35 mg/kg, s.c.) in both non-diabetic and diabetic mice. The selective kappa-opioid receptor antagonist nor-binaltorphimine (20 mg/kg, s.c.) also antagonized oxycodone-induced antinociception in diabetic mice, but only had a partial effect in non-diabetic mice. These results suggest that although primarily interacts with mu(1)-opioid receptor, kappa-opioid receptors are also strongly involved in oxycodone-induced antinociception.


Subject(s)
Analgesics, Opioid/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Oxycodone/pharmacology , Receptors, Opioid, mu/physiology , Animals , Male , Mice , Mice, Inbred ICR , Naloxone/analogs & derivatives , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Receptors, Opioid, mu/classification , Streptozocin
14.
J Pharmacol Sci ; 102(4): 377-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130673

ABSTRACT

The most consistent behavioral change caused by olfactory bulbectomy (OBX) is a hyperemotional response to novel environmental stimuli. The aim of this study was to characterize the emotional behavior of OBX mice using the hole-board test. After the olfactory bulbs were lesioned, sham and OBX mice were housed in single cages for 14 days. The number of head-dips in the hole-board test in single-housed OBX mice was significantly greater than that in single-housed sham mice. The head-dipping behaviors in single-housed sham and OBX mice were reversed by treatment with diazepam, a typical benzodiazepine anxiolytic. (+/-)-8-Hydroxy-2-(di-n-propylamino) tetraline hydrobromide (8-OH-DPAT), a selective 5-HT(1A)-receptor agonist that has a non-benzodiazepine anxiolytic-like effect, and (+)-4-[(aR)-a-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethyl benzamide (SNC80), a delta-opioid-receptor agonist, also significantly reversed the number of head-dips in single-housed sham and OBX mice. In conclusion, we suggest that the single-housed OBX mice showed heightened emotional behavior (e.g., increase in head-dipping behavior) in the hole-board test. In addition, we suggest that the hyperemotional behavior characterized by head-dipping behavior in OBX mice was selectively reversed by benzodiazepine and non-benzodiazepine anxiolytics.


Subject(s)
Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Emotions/drug effects , Exploratory Behavior/drug effects , Olfactory Bulb/surgery , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Benzamides/pharmacology , Diazepam/pharmacology , Dose-Response Relationship, Drug , Head Movements/drug effects , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Olfaction Disorders/chemically induced , Olfaction Disorders/psychology , Piperazines/pharmacology , Reaction Time/drug effects , Social Isolation , Zinc Sulfate
15.
Eur J Pharmacol ; 552(1-3): 99-104, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17056034

ABSTRACT

The involvement of opioid receptor activation in the antinociceptive effect of either fluvoxamine, a selective serotonin reuptake inhibitor, or serotonin (5-HT) on thermal hyperalgesia and mechanical allodynia in a model of neuropathic pain in mice induced by sciatic nerve ligation was examined. The experiments were conducted 2 or 6 weeks after unilateral sciatic nerve ligation. Ipsilateral thermal hyperalgesia and mechanical allodynia were observed both 2 and 6 weeks after sciatic nerve ligation. Neither s.c. fluvoxamine nor i.t. 5-HT affected sciatic nerve ligation-induced thermal hyperalgesia or mechanical allodynia in mice 2 weeks after sciatic nerve ligation. However, the same dose of either fluvoxamine or 5-HT significantly reduced mechanical allodynia but not thermal hyperalgesia in sciatic nerve ligated mice 6 weeks after surgery. The antinociceptive effect of fluvoxamine on sciatic nerve ligation-induced mechanical allodynia in mice 6 weeks after surgery was completely abolished by pretreatment with either naloxone, a nonselective opioid receptor antagonist, or beta-funaltrexamine, a selective mu-opioid receptor antagonist. Furthermore, pretreatment with naltrindole, a selective delta-opioid receptor antagonist, partially but significantly inhibited the antinociceptive effect of fluvoxamine in sciatic nerve ligated mice at the 6th postoperative week. The antinociceptive effect induced by i.t. 5-HT was also completely antagonized by either naloxone or beta-funaltrexamine, and partially inhibited by naltrindole. However, pretreatment with nor-binaltorphimine, a selective kappa-opioid receptor antagonist, had no effect against either s.c. fluvoxamine- or i.t. 5-HT-induced antinociception. These results suggest that the antinociceptive effect of s.c. fluvoxamine or i.t. 5-HT in the chronic state of sciatic nerve ligation-induced neuropathic pain may be related to opioidergic activity, mainly through the activation of spinal mu- and delta-opioid receptors.


Subject(s)
Analgesics/pharmacology , Fluvoxamine/pharmacology , Neuralgia/prevention & control , Receptors, Opioid/physiology , Sciatic Nerve/physiopathology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Dose-Response Relationship, Drug , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Injections, Spinal , Ketanserin/pharmacology , Male , Mice , Mice, Inbred ICR , Naloxone/pharmacology , Narcotic Antagonists , Neuralgia/physiopathology , Ondansetron/pharmacology , Sciatic Nerve/injuries , Sciatic Nerve/surgery , Serotonin/administration & dosage , Serotonin/pharmacology , Serotonin Agents/pharmacology , Serotonin Antagonists/pharmacology , Time Factors
16.
Eur J Pharmacol ; 535(1-3): 145-51, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16533506

ABSTRACT

We investigated the antinociceptive efficacy of systemic and centrally injected oxycodone on thermal hyperalgesia in streptozotocin-induced diabetic mice. The antinociceptive response was assessed by recording the latency in the tail-flick test using the radiant heat from a 50-W projection bulb on the tail. The tail-flick latency in diabetic mice was significantly shorter than that in non-diabetic mice. Oral (p.o.) and i.t., but not i.c.v., administration of oxycodone prolonged the tail-flick latency in diabetic mice to a level that was considerably longer than the baseline latency in non-diabetic mice. However, morphine did not significantly inhibit the tail-flick response in diabetic mice. The antinociceptive effect of either p.o. or i.t. oxycodone in non-diabetic mice, but not in diabetic mice, was antagonized by pretreatment with a selective mu-opioid receptor antagonist, beta-funaltrexamine. In non-diabetic mice, pretreatment with a selective kappa-opioid receptor antagonist, nor-binaltorphimine, had no effect on the peak antinociceptive effect of either p.o. or i.t. oxycodone at 30 min after administration, however, it slightly but significantly reduced oxycodone-induced antinociception at 60 and 90 min after administration. On the other hand, pretreatment with nor-binaltorphimine practically abolished the antinociceptive effects of both p.o.- and i.t.-administered oxycodone in diabetic mice. Naltrindole, a selective delta-opioid receptor antagonist, had no effects on the antinociceptive effect of oxycodone in either non-diabetic or diabetic mice. These results suggest that the antinociceptive effects of oxycodone may be mediated by spinal kappa-opioid receptors in diabetic mice, whereas it may interact primarily with supraspinal and spinal mu-opioid receptors in non-diabetic mice.


Subject(s)
Analgesics, Opioid/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Oxycodone/pharmacology , Pain/prevention & control , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Area Under Curve , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Injections, Intraventricular , Injections, Spinal , Injections, Subcutaneous , Mice , Mice, Inbred ICR , Naltrexone/administration & dosage , Naltrexone/analogs & derivatives , Naltrexone/pharmacokinetics , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacokinetics , Narcotic Antagonists/pharmacology , Oxycodone/administration & dosage , Oxycodone/pharmacokinetics , Pain/physiopathology , Pain Measurement/methods , Streptozocin
17.
Eur J Pharmacol ; 536(1-2): 123-7, 2006 Apr 24.
Article in English | MEDLINE | ID: mdl-16556439

ABSTRACT

In the present study, we examined the effect of mexiletine on vincristine-induced thermal hyperalgesia in mice. Mice were intraperitoneally treated with vincristine at a dose of 0.05 mg/kg one day after the measurement of the pre-drug latency in the tail-flick test, and then treated with a dose of 0.125 mg/kg twice a week for 6 weeks. In vincristine-treated mice, a significant decrease in tail-flick latency developed at 6 weeks after treatment. Pretreatment with mexiletine, at doses of 3, 10 and 30 mg/kg, i.p., dose-dependently increased the tail-flick latency in vincristine-treated mice. A significant reduction of the tail-flick latency was observed when the tail-flick latency was examined 60 min after i.t. administration of NG-nitro-L-arginine methyl ester (L-NAME, 30 nmol), a nitric oxide synthase (NOS) inhibitor, in naive mice. This L-NAME-induced thermal hyperalgesia was dose-dependently attenuated by pretreatment with mexiletine (10 and 30 mg/kg, i.p.), 10 min before the injection of L-NAME. The duration of nociceptive behavioral response induced by fenvalerate, at a dose of 0.1 microg, i.t., was significantly increased by pretreatment with L-NAME (30 nmol, i.t.). Intrathecal pretreatment with L-arginine (300 pmol) significantly reversed the L-NAME-induced enhancement of fenvalerate-induced nociceptive responses. The present study demonstrates that systemic mexiletine can effectively attenuate vincristine-induced thermal hyperalgesia. Furthermore, these results suggest that blockade of nitric oxide-induced enhancement of nociceptive transmission, in which tetrodotoxin-resistant sodium channels play an important role, may participate in the antinociceptive effect of mexiletine on vincristine-induced thermal hyperalgesia.


Subject(s)
Mexiletine/pharmacology , Neuralgia/prevention & control , Vincristine/toxicity , Animals , Arginine/administration & dosage , Arginine/pharmacology , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Injections, Intraperitoneal , Injections, Spinal , Male , Mexiletine/administration & dosage , Mice , Mice, Inbred ICR , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/toxicity , Neuralgia/chemically induced , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitriles/administration & dosage , Nitriles/toxicity , Pain Measurement/methods , Pyrethrins/administration & dosage , Pyrethrins/toxicity , Vincristine/administration & dosage
18.
Eur J Pharmacol ; 524(1-3): 75-9, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16256106

ABSTRACT

The effect of oxycodone on thermal hyperalgesia in streptozotocin-induced diabetic mice was examined. The antinociceptive response was assessed by recording the latency in the tail-flick test using the radiant heat from a 50-W projection bulb on the tail. The tail-flick latency in diabetic mice was significantly shorter than that in non-diabetic mice. When diabetic mice were treated with oxycodone (5 mg/kg, s.c.), the tail-flick latency in diabetic mice was prolonged to the level considerably longer than the baseline latencies of non-diabetic mice. However, s.c. administration of morphine (5 mg/kg) did not produce a significant inhibition of the tail-flick response in diabetic mice. Oxycodone, at doses of 1.25-5.0 mg/kg administered s.c., produced a dose-dependent increase in the tail-flick latencies in both diabetic and non-diabetic mice. The antinociceptive effect of oxycodone was antagonized by pretreatment with a selective delta-opioid receptor antagonist, beta-funaltrexamine (20 mg/kg, s.c.), in both non-diabetic and diabetic mice. In non-diabetic mice, pretreatment with a selective kappa-opioid receptor antagonist, nor-binaltorphimine (20 mg/kg, s.c.) had no effect on the peak antinociceptive effect of oxycodone observed 30 min after administration, however, it slightly but significantly reduced oxycodone-induced antinociception observed 60 and 90 min after administration. On the other hand, pretreatment with nor-binaltorphimine practically abolished the peak (30 min) and persistent (60 and 90 min) antinociceptive effects of oxycodone in diabetic mice. Naltrindole (35 mg/kg, s.c.), a selective delta-opioid receptor antagonist, had no effects on the antinociceptive effect of oxycodone in both non-diabetic and diabetic mice. These results suggest that the antinociceptive effects of oxycodone may be mediated by mu- and kappa-opioid receptors in diabetic mice, whereas it may interact primarily with mu-opioid receptors in non-diabetic mice.


Subject(s)
Analgesics, Opioid/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Oxycodone/pharmacology , Pain/prevention & control , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred ICR , Morphine/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Pain/physiopathology , Pain Measurement/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL