Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Toxics ; 11(12)2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38133405

ABSTRACT

Blood biomarkers, including neurofilament light chain (NfL), have garnered attention as potential indicators for chemotherapy-induced peripheral neuropathy (CIPN), a dose-limiting adverse effect of neurotoxic anticancer drugs. However, no blood biomarker has been established for routine application or translational research. This pilot study aimed to evaluate a limited panel of blood biomarkers in rat models of CIPN and their correlations with neuropathic pain. CIPN models were induced through repeated injections of oxaliplatin, paclitaxel, bortezomib, and vincristine. Electronic von Frey testing was used to assess tactile allodynia. Post anticancer injections, serum concentrations of 31 proteins were measured. Allodynia thresholds decreased in anticancer-treated animals compared to controls. No consistent modifications were observed in the biomarkers across CIPN models. The most noteworthy biomarkers with increased concentrations in at least two CIPN models were NfL (paclitaxel, vincristine), MCP-1, and RANTES (oxaliplatin, vincristine). Vincristine-treated animals exhibited strong correlations between LIX, MCP-1, NfL, and VEGF concentrations and tactile allodynia thresholds. No single biomarker can be recommended as a unique indicator of CIPN-related pain. Because of the study limitations (single dose of each anticancer drug, young animals, and single time measurement of biomarkers), further investigations are necessary to define the kinetics, specificities, and sensitivities of MCP-1, RANTES, and NfL.

2.
Biomolecules ; 13(2)2023 02 15.
Article in English | MEDLINE | ID: mdl-36830733

ABSTRACT

Diabetic neuropathy is often associated with chronic pain. Serotonin type 6 (5-HT6) receptor ligands, particularly inverse agonists, have strong analgesic potential and may be new candidates for treating diabetic neuropathic pain and associated co-morbid cognitive deficits. The current study addressed the involvement of 5-HT6 receptor constitutive activity and mTOR signaling in an experimental model of diabetic neuropathic pain induced by streptozocin (STZ) injection in the rat. Here, we show that mechanical hyperalgesia and associated cognitive deficits are suppressed by the administration of 5-HT6 receptor inverse agonists or rapamycin. The 5-HT6 receptor ligands also reduced tactile allodynia in traumatic and toxic neuropathic pain induced by spinal nerve ligation and oxaliplatin injection. Furthermore, both painful and co-morbid cognitive symptoms in diabetic rats are reduced by intrathecal delivery of a cell-penetrating peptide that disrupts 5-HT6 receptor-mTOR physical interaction. These findings demonstrate the deleterious influence of the constitutive activity of spinal 5-HT6 receptors upon painful and cognitive symptoms in diabetic neuropathic pains of different etiologies. They suggest that targeting the constitutive activity of 5-HT6 receptors with inverse agonists or disrupting the 5-HT6 receptor-mTOR interaction might be valuable strategies for the alleviation of diabetic neuropathic pain and cognitive co-morbidities.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Neuralgia , Rats , Animals , Diabetes Mellitus, Experimental/complications , Drug Inverse Agonism , Ligands , Serotonin/pharmacology , Hyperalgesia , TOR Serine-Threonine Kinases
3.
Biomed Pharmacother ; 149: 112915, 2022 May.
Article in English | MEDLINE | ID: mdl-35635358

ABSTRACT

BACKGROUND: Donepezil, a cholinesterase inhibitor approved in Alzheimer's disease, has demonstrated analgesic and preventive effects in animal models of oxaliplatin-induced neuropathy. To improve the clinical interest of donepezil for the management and prevention of chemotherapy-induced peripheral neuropathy (CIPN), a broader validation is required in different animal models of CIPN. METHODS: using rat models of CIPN (bortezomib, paclitaxel, and vincristine), the analgesic and preventive efficacies of donepezil were evaluated on tactile, cold and heat hypersensitivities. The involvement of muscarinic M2 acetylcholine receptors (m2AChRs) in analgesic effects was investigated at the spinal level. The absence of interference of donepezil with the cytotoxic effect of chemotherapy has been controlled in cancer cell lines. RESULTS: the analgesic efficacy of donepezil was demonstrated for all CIPN models, mainly on tactile hypersensitivity (maximal efficacy at 60 min, p < 0.05 vs. vehicle group). This effect was suppressed by an intrathecal injection of methoctramine (m2AChR antagonist). Regarding preventive effects, donepezil limited tactile hypersensitivity induced by paclitaxel, but not for other CIPN models. Donepezil did not modify the viability of cancer cells or the efficacy of anticancer drugs. CONCLUSIONS: donepezil had a broad analgesic effect on animal models of CIPN and this effect involved spinal m2AChRs. This work validates the repositioning of donepezil in the management of CIPN.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Peripheral Nervous System Diseases , Acetylcholine , Analgesics/adverse effects , Animals , Antineoplastic Agents/toxicity , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Donepezil , Models, Animal , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control , Rats , Receptor, Muscarinic M2 , Receptors, Muscarinic
4.
Br J Pharmacol ; 177(20): 4782-4795, 2020 10.
Article in English | MEDLINE | ID: mdl-32851651

ABSTRACT

BACKGROUND AND PURPOSE: Opioids are effective painkillers. However, their risk-benefit ratio is dampened by numerous adverse effects and opioid misuse has led to a public health crisis. Safer alternatives are required, but isolating the antinociceptive effect of opioids from their adverse effects is a pharmacological challenge because activation of the µ opioid receptor triggers both the antinociceptive and adverse effects of opioids. EXPERIMENTAL APPROACH: The TREK1 potassium channel is activated downstream of µ receptor and involved in the antinociceptive activity of morphine but not in its adverse effects. Bypassing the µ opioid receptor to directly activate TREK1 could therefore be a safer analgesic strategy. KEY RESULTS: We developed a selective TREK1 activator, RNE28, with antinociceptive activity in naive rodents and in models of inflammatory and neuropathic pain. This activity was lost in TREK1 knockout mice or wild-type mice treated with the TREK1 blocker spadin, showing that TREK1 is required for the antinociceptive activity of RNE28. RNE28 did not induce respiratory depression, constipation, rewarding effects, or sedation at the analgesic doses tested. CONCLUSION AND IMPLICATIONS: This proof-of-concept study shows that TREK1 activators could constitute a novel class of painkillers, inspired by the mechanism of action of opioids but devoid of their adverse effects.


Subject(s)
Analgesics, Opioid , Neuralgia , Analgesics , Analgesics, Opioid/adverse effects , Animals , Mice , Mice, Knockout , Morphine , Receptors, Opioid, mu
5.
Prog Neurobiol ; 193: 101846, 2020 10.
Article in English | MEDLINE | ID: mdl-32512114

ABSTRACT

Chronic neuropathic pain is a highly disabling syndrome that is poorly controlled by currently available analgesics. Here, we show that painful symptoms and associated cognitive deficits induced by spinal nerve ligation in the rat are prevented by the administration of serotonin 5-HT6 receptor inverse agonists or by the mTOR inhibitor rapamycin. In contrast, they are not alleviated by the administration of 5-HT6 receptor neutral antagonists. Likewise, activation of mTOR by constitutively active 5-HT6 receptors mediates allodynia in oxaliplatin-induced peripheral neuropathy in rats but not mechanical nociception in healthy rats. Furthermore, both painful and co-morbid cognitive symptoms in neuropathic rats are strongly reduced by intrathecal delivery of a cell-penetrating peptide that disrupts 5-HT6 receptor/mTOR physical interaction. Collectively, these findings demonstrate a deleterious influence of non-physiological mTOR activation by constitutively active spinal 5-HT6 receptors upon painful and cognitive symptoms in neuropathic pains of different etiologies. They suggest that targeting the constitutive activity of 5-HT6 receptors with inverse agonists or disrupting the 5-HT6 receptor/mTOR interaction might be valuable strategies for the alleviation of neuropathic pain and cognitive co-morbidities.


Subject(s)
Cognitive Dysfunction , Hyperalgesia , Neuralgia , Nociception , Receptors, Serotonin , Serotonin Agents/pharmacology , TOR Serine-Threonine Kinases , Animals , Behavior, Animal/drug effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuralgia/complications , Neuralgia/drug therapy , Neuralgia/metabolism , Nociception/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Serotonin Agents/administration & dosage , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
6.
FASEB J ; 34(6): 7483-7499, 2020 06.
Article in English | MEDLINE | ID: mdl-32277850

ABSTRACT

Recent studies have revealed gender differences in cold perception, and pointed to a possible direct action of testosterone (TST) on the cold-activated TRPM8 (Transient Receptor Potential Melastatin Member 8) channel. However, the mechanisms by which TST influences TRPM8-mediated sensory functions remain elusive. Here, we show that TST inhibits TRPM8-mediated mild-cold perception through the noncanonical engagement of the Androgen Receptor (AR). Castration of both male rats and mice increases sensitivity to mild cold, and this effect depends on the presence of intact TRPM8 and AR. TST in nanomolar concentrations suppresses whole-cell TRPM8-mediated currents and single-channel activity in native dorsal root ganglion (DRG) neurons and HEK293 cells co-expressing recombinant TRPM8 and AR, but not TRPM8 alone. AR cloned from rat DRGs shows no difference from standard AR. However, biochemical assays and confocal imaging reveal the presence of AR on the cell surface and its interaction with TRPM8 in response to TST, leading to an inhibition of channel activity.


Subject(s)
Receptors, Androgen/metabolism , TRPM Cation Channels/metabolism , Testosterone/metabolism , Androgens/metabolism , Animals , Cell Line , Cold Temperature , Female , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Rats , Rats, Wistar
7.
Neuropharmacology ; 125: 308-318, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28780039

ABSTRACT

Antidepressants remain one of the first line treatments prescribed to neuropathic pain patients despite their limited efficacy and/or their numerous side effects. More and more, pharmacotherapy for neuropathic pain has evolved towards the use of therapeutic combinations. The goal of the present study was to assess the efficacy of the combination of antidepressants - selective serotonin reuptake inhibitors and serotonin-noradrenaline reuptake inhibitors-with a peptide (TAT-2ASCV) able to disrupt the interaction between serotonin type 2A (5-HT2A) receptors and associated PDZ proteins. Mechanical hypersensitivity was assessed in sciatic nerve ligation-induced neuropathic pain in rats using paw pressure test after acute treatment with TAT-2ASCV alone or in combination with repeated treatment with fluoxetine or duloxetine or clomipramine. First, we validated the anti-hyperalgesic effect of TAT-2ASCV on mechanical hypersensitivity at the dose of 100 ng/rat (single i.t. injection). Second, using selective receptor antagonists, we found that the effect of TAT-2ASCV on mechanical hypersensitivity involves 5-HT2A as well as GABAA receptors. Finally, we showed that the association of TAT-2ASCV (100 ng, single i.t. injection) with fluoxetine (10 mg/kg, five i.p. injections) reveals its anti-hyperalgesic effect, while the association with duloxetine (1 mg/kg, five i.p. injections) or clomipramine (2.5 mg/kg, five i.p. injections) is only additive. Those results further accentuate the interest to develop small molecules acting like TAT-2ASCV in order to treat neuropathic pain as a monotherapy or in combination with antidepressants.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Neuralgia/drug therapy , PDZ Domains , Receptor, Serotonin, 5-HT2A/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Animals , Clomipramine/pharmacology , Disease Models, Animal , Duloxetine Hydrochloride/pharmacology , Fluoxetine/pharmacology , GABA-A Receptor Agonists/pharmacology , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Neuralgia/metabolism , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Sciatic Nerve , Serotonin 5-HT2 Receptor Agonists/pharmacology , Touch
8.
J Med Chem ; 60(3): 1076-1088, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28051863

ABSTRACT

The TWIK-related K+ channel, TREK-1, has recently emerged as an attractive therapeutic target for the development of a novel class of analgesic drugs, suggesting that activation of TREK-1 could result in pain inhibition. Here, we report the synthesis of a series of substituted acrylic acids (1-54) based on our previous work with caffeate esters. The analogues were evaluated for their ability to modulate TREK-1 channel by electrophysiology and for their in vivo antinociceptive activity (acetic acid-induced writhing and hot plate assays), leading to the identification of a series of novel molecules able to activate TREK-1 and displaying potent antinociceptive activity in vivo. Furyl analogue 36 is the most promising of the series.


Subject(s)
Analgesics/pharmacology , Potassium Channels, Tandem Pore Domain/agonists , Animals
9.
Pain ; 158(1): 149-160, 2017 01.
Article in English | MEDLINE | ID: mdl-27984527

ABSTRACT

Antidepressants are first-line treatments of neuropathic pain but not all these drugs are really effective. Agomelatine is an antidepressant with a novel mode of action, acting as an MT1/MT2 melatonergic receptor agonist and a 5-HT2C receptor antagonist that involves indirect norepinephrine release. Melatonin, serotonin, and norepinephrine have been involved in the pathophysiology of neuropathic pain. Yet, no study has been conducted to determine agomelatine effects on neuropathic pain in animal models. Using 3 rat models of neuropathic pain of toxic (oxaliplatin/OXA), metabolic (streptozocin/STZ), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, we investigated the antihypersensitivity effect of acute and repeated agomelatine administration. We then determined the influence of melatonergic, 5-HT2C, α-2 and ß-1/2 adrenergic receptor antagonists in the antihypersensitivity effect of agomelatine. The effect of the combination of agomelatine + gabapentin was evaluated using an isobolographic approach. In STZ and CCI models, single doses of agomelatine significantly and dose dependently reduced mechanical hypersensitivity. After daily administrations for 2 weeks, this effect was confirmed in the CCI model and agomelatine also displayed a marked antihypersensitivity effect in the OXA model. The antihypersensitivity effect of agomelatine involved melatonergic, 5-HT2C, and α-2 adrenergic receptors but not beta adrenoceptors. The isobolographic analysis demonstrated that the combination of agomelatine + gabapentin had additive effects. Agomelatine exerts a clear-cut antihypersensitivity effect in 3 different neuropathic pain models. Its effect is mediated by melatonergic and 5-HT2C receptors and, although agomelatine has no affinity, also by α-2 adrenergic receptors. Finally, agomelatine combined with gabapentin produces an additive antihypersensitivity effect.


Subject(s)
Acetamides/therapeutic use , Hypnotics and Sedatives/therapeutic use , Neuralgia/drug therapy , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Amines/therapeutic use , Animals , Antineoplastic Agents/toxicity , Constriction, Pathologic/complications , Cyclohexanecarboxylic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Gabapentin , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Idazoxan/therapeutic use , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuralgia/etiology , Organoplatinum Compounds/toxicity , Oxaliplatin , Pain Measurement , Rats , Rats, Sprague-Dawley , Thiophenes/therapeutic use , gamma-Aminobutyric Acid/therapeutic use
10.
Cell Rep ; 11(7): 1067-78, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25959819

ABSTRACT

Cold-triggered pain is essential to avoid prolonged exposure to harmfully low temperatures. However, the molecular basis of noxious cold sensing in mammals is still not completely understood. Here, we show that the voltage-gated Nav1.9 sodium channel is important for the perception of pain in response to noxious cold. Nav1.9 activity is upregulated in a subpopulation of damage-sensing sensory neurons responding to cooling, which allows the channel to amplify subthreshold depolarizations generated by the activation of cold transducers. Consequently, cold-triggered firing is impaired in Nav1.9(-/-) neurons, and Nav1.9 null mice and knockdown rats show increased cold pain thresholds. Disrupting Nav1.9 expression in rodents also alleviates cold pain hypersensitivity induced by the antineoplastic agent oxaliplatin. We conclude that Nav1.9 acts as a subthreshold amplifier in cold-sensitive nociceptive neurons and is required for the perception of cold pain under normal and pathological conditions.


Subject(s)
Hyperalgesia/metabolism , NAV1.9 Voltage-Gated Sodium Channel/metabolism , Pain Perception/physiology , Thermosensing/physiology , Animals , Cold Temperature , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
11.
Eur J Med Chem ; 75: 391-402, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24561669

ABSTRACT

The TWIK-related K(+) channel, TREK-1, has recently emerged as an attractive therapeutic target for the development of a novel class of analgesic drugs. It has been reported that TREK-1 -/- mice were more sensitive than wild-type mice to painful stimuli, suggesting that activation of TREK-1 could result in pain inhibition. Here we report the synthesis of a series of substituted caffeate esters (12a-u) based on the hit compound CDC 2 (cinnamyl 3,4-dihydroxyl-α-cyanocinnamate). These analogs were evaluated for their ability to modulate TREK-1 channel by electrophysiology and for their in vivo antinociceptive activity (acetic acid induced-writhing assay) leading to the identification a series of novel molecules able to activate TREK-1 and displaying potent analgesic activity in vivo.


Subject(s)
Analgesics/chemistry , Analgesics/therapeutic use , Caffeic Acids/chemistry , Caffeic Acids/therapeutic use , Pain/drug therapy , Potassium Channels, Tandem Pore Domain/metabolism , Analgesics/pharmacology , Animals , Caffeic Acids/pharmacology , Cinnamates/chemistry , Cinnamates/pharmacology , Cinnamates/therapeutic use , Esters/chemistry , Esters/pharmacology , Esters/therapeutic use , Male , Mice , Models, Molecular , Quantitative Structure-Activity Relationship , Xenopus
12.
J Neurosci ; 33(48): 18951-65, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24285900

ABSTRACT

Hyperactivity of the glutamatergic system is involved in the development of central sensitization in the pain neuraxis, associated with allodynia and hyperalgesia observed in patients with chronic pain. Herein we study the ability of type 4 metabotropic glutamate receptors (mGlu4) to regulate spinal glutamate signaling and alleviate chronic pain. We show that mGlu4 are located both on unmyelinated C-fibers and spinal neurons terminals in the inner lamina II of the spinal cord where they inhibit glutamatergic transmission through coupling to Cav2.2 channels. Genetic deletion of mGlu4 in mice alters sensitivity to strong noxious mechanical compression and accelerates the onset of the nociceptive behavior in the inflammatory phase of the formalin test. However, responses to punctate mechanical stimulation and nocifensive responses to thermal noxious stimuli are not modified. Accordingly, pharmacological activation of mGlu4 inhibits mechanical hypersensitivity in animal models of inflammatory or neuropathic pain while leaving acute mechanical perception unchanged in naive animals. Together, these results reveal that mGlu4 is a promising new target for the treatment of chronic pain.


Subject(s)
Excitatory Amino Acid Agonists/therapeutic use , Hyperalgesia/drug therapy , Receptors, Metabotropic Glutamate/agonists , Animals , Blotting, Western , Carrageenan , Chronic Disease , Constriction, Pathologic/pathology , Electrophysiological Phenomena/physiology , Fluorescent Antibody Technique , Immersion/physiopathology , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Myelin Sheath/physiology , Pain Measurement/drug effects , Patch-Clamp Techniques , Phosphinic Acids/administration & dosage , Phosphinic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics , Rhizotomy , Sensory Receptor Cells/physiology , Spinal Cord/cytology , Spinal Cord/physiology , Synaptic Transmission/physiology
13.
PLoS One ; 8(8): e70690, 2013.
Article in English | MEDLINE | ID: mdl-23940628

ABSTRACT

The discovery that paracetamol is metabolized to the potent TRPV1 activator N-(4-hydroxyphenyl)-5Z,8Z,11Z,14Z-eicosatetraenamide (AM404) and that this metabolite contributes to paracetamol's antinociceptive effect in rodents via activation of TRPV1 in the central nervous system (CNS) has provided a potential strategy for developing novel analgesics. Here we validated this strategy by examining the metabolism and antinociceptive activity of the de-acetylated paracetamol metabolite 4-aminophenol and 4-hydroxy-3-methoxybenzylamine (HMBA), both of which may undergo a fatty acid amide hydrolase (FAAH)-dependent biotransformation to potent TRPV1 activators in the brain. Systemic administration of 4-aminophenol and HMBA led to a dose-dependent formation of AM404 plus N-(4-hydroxyphenyl)-9Z-octadecenamide (HPODA) and arvanil plus olvanil in the mouse brain, respectively. The order of potency of these lipid metabolites as TRPV1 activators was arvanil = olvanil>>AM404> HPODA. Both 4-aminophenol and HMBA displayed antinociceptive activity in various rodent pain tests. The formation of AM404, arvanil and olvanil, but not HPODA, and the antinociceptive effects of 4-aminophenol and HMBA were substantially reduced or disappeared in FAAH null mice. The activity of 4-aminophenol in the mouse formalin, von Frey and tail immersion tests was also lost in TRPV1 null mice. Intracerebroventricular injection of the TRPV1 blocker capsazepine eliminated the antinociceptive effects of 4-aminophenol and HMBA in the mouse formalin test. In the rat, pharmacological inhibition of FAAH, TRPV1, cannabinoid CB1 receptors and spinal 5-HT3 or 5-HT1A receptors, and chemical deletion of bulbospinal serotonergic pathways prevented the antinociceptive action of 4-aminophenol. Thus, the pharmacological profile of 4-aminophenol was identical to that previously reported for paracetamol, supporting our suggestion that this drug metabolite contributes to paracetamol's analgesic activity via activation of bulbospinal pathways. Our findings demonstrate that it is possible to construct novel antinociceptive drugs based on fatty acid conjugation as a metabolic pathway for the generation of TRPV1 modulators in the CNS.


Subject(s)
Amidohydrolases/metabolism , Aminophenols/pharmacology , Analgesics/pharmacology , Benzylamines/pharmacology , TRPV Cation Channels/agonists , Aminophenols/pharmacokinetics , Analgesics/pharmacokinetics , Animals , Arachidonic Acids/metabolism , Benzylamines/pharmacokinetics , Brain/metabolism , Capsaicin/analogs & derivatives , Capsaicin/metabolism , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Nociception/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , TRPV Cation Channels/metabolism , Vasodilation/drug effects
14.
Pain ; 153(8): 1657-1663, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22664319

ABSTRACT

Chronic pain is a multidimensional experience that not only includes changes in nociception, but also impairments in emotion and cognitive functions. These last 2 components are not often taken into account in preclinical research. We investigated emotional and cognitive impairments in a model of neuropathic pain in rats induced by chronic constriction injury (CCI) of the sciatic nerve. Nociceptive response, anxiety and depressive-like behaviours as well as cognitive capacities were analysed, and the effect of per os administration of duloxetine and gabapentin was studied. In the electronic von Frey test, CCI rats exhibited mechanical hypersensitivity which can be influenced by duloxetine (3-30 mg/kg) and gabapentin (10-30 mg/kg). Cognitive impairments were found in the social but not in the spatial (Y-maze) recognition memory tests. Duloxetine and gabapentin dose-dependently (3-30 mg/kg) restored social recognition memory impairment. Anxiety-like behaviour was only observed in the open-field test (decrease in the time spent in the inner zone) but not in the elevated plus maze or in the social interactions tests in CCI animals. In this test, impairment in locomotor activity (decrease of the total number of crossing) was also observed. Duloxetine and gabapentin (10mg/kg) were effective to increase the time spent in the inner zone as well as locomotor activity. No difference was observed in depressive-like behaviour (saccharin preference test) between sham-operated and CCI rats. These data suggest that cognitive rather than emotional impairments seem to be present in neuropathic CCI rats and can be reversed by duloxetine and gabapentin.


Subject(s)
Affective Symptoms/drug therapy , Amines/pharmacology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cyclohexanecarboxylic Acids/pharmacology , Neuralgia/drug therapy , Neuralgia/physiopathology , Thiophenes/pharmacology , gamma-Aminobutyric Acid/pharmacology , Affective Symptoms/etiology , Affective Symptoms/physiopathology , Analgesics/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Cognition/drug effects , Cognition Disorders/etiology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Duloxetine Hydrochloride , Emotions/drug effects , Gabapentin , Male , Neuralgia/complications , Rats , Rats, Sprague-Dawley , Treatment Outcome
15.
PLoS One ; 6(8): e23083, 2011.
Article in English | MEDLINE | ID: mdl-21857998

ABSTRACT

Inflammation is known to be responsible for the sensitization of peripheral sensory neurons, leading to spontaneous pain and invalidating pain hypersensitivity. Given its role in regulating neuronal excitability, the voltage-gated Nav1.9 channel is a potential target for the treatment of pathological pain, but its implication in inflammatory pain is yet not fully described. In the present study, we examined the role of the Nav1.9 channel in acute, subacute and chronic inflammatory pain using Nav1.9-null mice and Nav1.9 knock-down rats. In mice we found that, although the Nav1.9 channel does not contribute to basal pain thresholds, it plays an important role in heat pain hypersensitivity induced by subacute paw inflammation (intraplantar carrageenan) and chronic ankle inflammation (complete Freund's adjuvant-induced monoarthritis). We showed for the first time that Nav1.9 also contributes to mechanical hypersensitivity in both models, as assessed using von Frey and dynamic weight bearing tests. Consistently, antisense-based Nav1.9 gene silencing in rats reduced carrageenan-induced heat and mechanical pain hypersensitivity. While no changes in Nav1.9 mRNA levels were detected in dorsal root ganglia (DRGs) during subacute and chronic inflammation, a significant increase in Nav1.9 immunoreactivity was observed in ipsilateral DRGs 24 hours following carrageenan injection. This was correlated with an increase in Nav1.9 immunolabeling in nerve fibers surrounding the inflamed area. No change in Nav1.9 current density could be detected in the soma of retrolabeled DRG neurons innervating inflamed tissues, suggesting that newly produced channels may be non-functional at this level and rather contribute to the observed increase in axonal transport. Our results provide evidence that Nav1.9 plays a crucial role in the generation of heat and mechanical pain hypersensitivity, both in subacute and chronic inflammatory pain models, and bring new elements for the understanding of its regulation in those models.


Subject(s)
Hyperalgesia/physiopathology , Inflammation/physiopathology , Pain/physiopathology , Sodium Channels/physiology , Animals , Arthritis, Experimental/physiopathology , Carrageenan , Chronic Disease , Edema/chemically induced , Edema/physiopathology , Forelimb/drug effects , Forelimb/metabolism , Forelimb/physiopathology , Ganglia, Spinal/metabolism , Gene Expression , Gene Knockdown Techniques , Hindlimb/drug effects , Hindlimb/metabolism , Hindlimb/physiopathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.9 Voltage-Gated Sodium Channel , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sensory Receptor Cells/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Time Factors
16.
Pain ; 139(1): 190-200, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18485596

ABSTRACT

Acetaminophen is the most used analgesic/antipyretic drug. Its unclear mechanism of action could rely on cyclooxygenase inhibition, NO synthesis blockade or reinforcement of the serotonergic system. Here we show that in thermal, mechanical and chemical pain tests, AM-251, a specific CB(1) receptor antagonist, abolished the analgesic action of acetaminophen, which was also lost in CB(1) receptor knockout mice. Moreover, acetaminophen was shown unable to bind to CB(1) receptors demonstrating an indirect involvement of these receptors in the analgesic effect of this compound. Accordingly with these results, we also demonstrated that the inhibition of FAAH, an enzyme involved in the cerebral metabolism of acetaminophen into AM404, known to reinforce the activity of the endocannabinoid system, suppressed the antinociceptive effect of acetaminophen. In addition, similarly to the interaction of acetaminophen with bulbospinal serotonergic pathways and spinal serotonin receptors, we observed that the antinociceptive activity of ACEA, a CB(1) receptor agonist, was inhibited by lesion of bulbospinal serotonergic pathways and antagonists of spinal 5-HT receptors. We therefore propose that acetaminophen-induced analgesia could involve the following sequence: (1) FAAH-dependent metabolism of acetaminophen into AM404; (2) indirect involvement of CB(1) receptors by this metabolite; (3) endocannabinoid-dependent reinforcement of the serotonergic bulbospinal pathways, and (4) involvement of spinal pain-suppressing serotonergic receptors.


Subject(s)
Acetaminophen/therapeutic use , Analgesia/methods , Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Receptor, Cannabinoid, CB1/physiology , Serotonin/physiology , Acetaminophen/pharmacology , Animals , Cannabinoid Receptor Modulators/antagonists & inhibitors , Dose-Response Relationship, Drug , Male , Mice , Mice, Knockout , Pain/drug therapy , Pain/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, Serotonin/physiology , Serotonin/metabolism
17.
Pain ; 137(1): 112-124, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17900808

ABSTRACT

Glutamate plays a key role in modulation of nociceptive processing. This excitatory amino acid exerts its action through two distinct types of receptors, ionotropic and metabotropic glutamate receptors (mGluRs). Eight mGluRs have been identified and divided in three groups based on their sequence similarity, pharmacology and G-protein coupling. While the role of group I and II mGluRs is now well established, little is known about the part played by group III mGluRs in pain. In this work, we studied comparatively the involvement of spinal group III mGluR in modulation of acute, inflammatory and neuropathic pain. While intrathecal injection of ACPT-I, a selective group III mGluR agonist, failed to induce any change in vocalization thresholds of healthy animals submitted to mechanical or thermal stimuli, it dose-dependently inhibited the nociceptive behavior of rats submitted to the formalin test and the mechanical hyperalgesia associated with different animal models of inflammatory (carrageenan-treated and monoarthritic rats) or neuropathic pain (mononeuropathic and vincristine-treated rats). Similar effects were also observed following intrathecal injection of PHCCC, a positive allosteric modulator of mGlu4. Antihyperalgesia induced by ACPT-I was blocked either by LY341495, a nonselective antagonist of mGluR, by MAP4, a selective group III antagonist. This study provide new evidences supporting the role of spinal group III mGluRs in the modulation of pain perception in different pathological pain states of various etiologies but not in normal conditions. It more particularly highlights the specific involvement of mGlu4 in this process and may be a useful therapeutic approach to chronic pain treatment.


Subject(s)
Disease Models, Animal , Hyperalgesia/prevention & control , Neuralgia/prevention & control , Receptors, Metabotropic Glutamate/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Humans , Hyperalgesia/physiopathology , Inflammation/physiopathology , Inflammation/prevention & control , Male , Neuralgia/physiopathology , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists
18.
Mol Pharmacol ; 71(2): 407-15, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17088403

ABSTRACT

The mechanism of action of acetaminophen is currently widely discussed. Direct inhibition of cyclooxygenase isoforms remains the commonly advanced hypothesis. We combined behavioral studies with molecular techniques to investigate the mechanism of action of acetaminophen in a model of tonic pain in rats. We show that acetaminophen indirectly stimulates spinal 5-hydroxytryptamine (5-HT)1A receptors in the formalin test, thereby increasing transcript and protein levels of low-affinity neurotrophin receptor, insulin-like growth factor-1 (IGF-1) receptor alpha subunit, and growth hormone receptor and reducing the amount of somatostatin 3 receptor (sst3R) mRNA. Those cellular events seem to be important for the antinociceptive activity of acetaminophen. Indeed, down-regulation of sst3R mRNA depends on acetaminophen-elicited, 5-HT1A receptor-dependent increase in neuronal extracellular signal-regulated kinase 1/2 (ERK1/2) activities that mediate antinociception. In addition, spinal growth hormone (GH) and IGF-1 receptors would also be involved in the antinociceptive activity of the analgesic at different degrees. Our results show the involvement of specific 5-HT1A receptor-dependent cellular events in acetaminophen-produced antinociception and consequently indicate that inhibition of cyclooxygenase activities is not the exclusive mechanism involved. Furthermore, we propose that the mechanisms of 5-HT1A receptor-elicited antinociception and the role of the spinal ERK1/2 pathway in nociception are more intricate than suspected so far and that the GH/IGF-1 axis is an interesting new player in the regulation of spinal nociception.


Subject(s)
Acetaminophen/pharmacology , Analgesia , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Somatotropin/metabolism , Analgesics, Non-Narcotic/pharmacology , Animals , Cyclooxygenase Inhibitors , Pain/drug therapy , Pain/metabolism , Rats , Signal Transduction , Spine/chemistry , Spine/metabolism
19.
Eur J Pain ; 10(8): 749-55, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16427791

ABSTRACT

The number of old and very old persons is increasing and there is evidence that aging coincides with chronic painful conditions. Pain induces behavioural disorders that have been so far poorly identified in old and even less in very old animals. The aim of this study was to: (1) compare the evolution of pain in senescent animals (37-39 months) to old (20-22 months) and young (4-6 months) Lou/cjall rats after a chronic constriction of the sciatic nerve; (2) evaluate pain during four weeks after surgery with an experimental and an observational approach to determine how the response to noxious stimuli correlates with recorded spontaneous behaviour. Results showed that senescent animals are less sensitive to neuropathic pain than old or young rats while senescent/old rats are more sensitive to acute pain. The correlation between observational and experimental pain scores stresses the reliability of non-invasive measures for pain evaluation in senescent populations. The dichotomy between neuropathic and acute pain perceptions with age needs to be further investigated and would help to better understand the reasons of this uneven pain perception and expression with age.


Subject(s)
Age Factors , Behavior, Animal , Neuralgia/psychology , Acute Disease , Animals , Chronic Disease , Hyperalgesia/psychology , Male , Pain Measurement , Pain Threshold/psychology , Rats
20.
Gastroenterology ; 128(7): 1996-2008, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15940632

ABSTRACT

BACKGROUND & AIMS: The treatment of irritable bowel syndrome (IBS), characterized by abdominal pain and bloating, is empirical and often poorly efficient. Research lacks suitable models for studying the pathophysiologic mechanisms of the colonic hypersensitivity and new pharmacologic targets. The present study aimed to develop a novel model of colonic hypersensitivity possessing several of the characteristics encountered in patients with IBS. METHODS: Rats received enemas of a butyrate solution (8-1000 mmol/L) twice daily for 3 days. A time course was determined for colonic hypersensitivity (colorectal distention test) and referred cutaneous lumbar hyperalgesia (von Frey hairs). Macroscopic and histologic analyses were performed on colonic mucosa. The efficacy of morphine, U50488H (a kappa opioid agonist), and trimebutine on the 2 pain parameters was determined. Finally, the involvement of peptidergic C-fibers was evaluated using capsaicin-pretreated animals and treatments with calcitonin gene-related peptide (CGRP) and neurokinin 1 receptor antagonists. RESULTS: Butyrate enemas induced a sustained, concentration-dependent colonic hypersensitivity and, to a lesser extent, a referred cutaneous mechanical hyperalgesia, particularly in female rats, but no macroscopic and histologic modifications of the colonic mucosa, as observed in patients with IBS. Both pain parameters were sensitive to morphine, U50488H, trimebutine, neonatal capsaicin treatment, and the CGRP receptor antagonist but not to the neurokinin 1 receptor antagonist. CONCLUSIONS: These results present our noninflammatory model of chronic colonic hypersensitivity as a useful novel tool for studying IBS. The CGRP receptor antagonist-induced reduction of colonic hypersensitivity suggests that CGRP receptors may provide a promising target for treatment of IBS.


Subject(s)
Butyrates/adverse effects , Colonic Diseases/immunology , Disease Models, Animal , Irritable Bowel Syndrome/physiopathology , Receptors, Calcitonin Gene-Related Peptide/physiology , Animals , Butyrates/administration & dosage , Colonic Diseases/physiopathology , Dose-Response Relationship, Drug , Enema/veterinary , Female , Humans , Hyperalgesia/etiology , Hypersensitivity , Male , Pain/etiology , Rats , Rats, Sprague-Dawley , Rectum/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...