Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters










Publication year range
1.
J Physiol Pharmacol ; 70(6)2019 Dec.
Article in English | MEDLINE | ID: mdl-32203940

ABSTRACT

Streptozotocin (STZ) is commonly used to induce diabetes mellitus in experimental animal studies on peripheral diabetic neuropathy (PDN). Animals with STZ model of diabetes commonly develop changes in test stimulus-evoked pain behavior. However, it is still unclear whether rats with STZ model of diabetes have ongoing pain. Here we assessed whether STZ-induced diabetes induces ongoing pain-like behavior in male rats using conditioned place-preference (CPP) paradigm. CPP was tested in the fourth week of diabetes by pairing one chamber of the CPP device with vehicle and another chamber with either pregabalin (an established analgesic; 30 mg/kg i.p.; n = 9) or Chembridge-5861528 (a TRPA1 channel antagonist; 30 mg/kg i.p.; n = 9). After drug-pairings, the animals were allowed to choose which chamber they preferred. Mechanical sensitivity was assessed with monofilaments and chemonociception in the skin by determining mustard oil-induced pain behavior. Diabetic animals developed in two weeks mechanical hypersensitivity that changed into hyposensitivity by the fourth week. Mustard oil-induced sustained pain was reduced by the 4th week. After 4 weeks of diabetes, neither pregabalin nor the TRPA1 antagonist induced a significant overall change in the median CPP, although both drugs significantly reduced median withdrawal responses evoked by noxious mechanical stimulation. Pregabalin-induced CPP, however, had a significant positive correlation with the sustained pain behaviour induced by topical mustard oil. In conclusion, the present results suggest that the response to topical mustard oil may predict ongoing pain-like behavior in the STZ model of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/physiopathology , Pain/physiopathology , Analgesics/pharmacology , Animals , Conditioning, Psychological/physiology , Disease Models, Animal , Male , Mustard Plant/toxicity , Plant Oils/toxicity , Pregabalin/pharmacology , Rats , Rats, Wistar , Streptozocin , TRPA1 Cation Channel/antagonists & inhibitors
2.
Physiol Res ; 67(4): 647-655, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29750883

ABSTRACT

Here we studied whether descending control of mechanical nociception by glutamate in the central nucleus of the amygdala (CeA) of healthy control animals is induced by amygdaloid NMDA receptors and relayed through the midbrain periaqueductal gray (PAG). Mechanical nociception in the hind paws was assessed in rats with chronic guide cannulae for glutamate administration in the right CeA and for inducing local anesthesia in the PAG. In a separate electrophysiological study, ON-like PAG neurons giving an excitatory response to noxious pinch of the tail were recorded in anesthetized rats following glutamate administration into the CeA. A high dose of glutamate (100 microg) in the CeA induced mechanical antinociception in the contra- but not ipsilateral hind limb. Antinociception was prevented by an NMDA receptor antagonist in the CeA or local anesthesia of the PAG. Discharge rate of ON-like PAG neurons was increased by a high dose of glutamate (100 microg) in the CeA and this increase was prevented by an NMDA receptor antagonist in the CeA. The results indicate that amygdaloid NMDA receptors in the CeA may induce contralaterally mechanical antinociception through a circuitry relaying in the PAG. Activation of ON-like PAG neurons is associated with the descending antinociceptive effect. Mechanisms and causality of this association still remain to be studied.


Subject(s)
Central Amygdaloid Nucleus/physiology , Nerve Net/physiology , Pain Measurement/methods , Pain/physiopathology , Periaqueductal Gray/physiology , Animals , Central Amygdaloid Nucleus/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/toxicity , Male , Nerve Net/drug effects , Pain/chemically induced , Pain/prevention & control , Pain Measurement/drug effects , Periaqueductal Gray/drug effects , Rats , Rats, Wistar
3.
Neuroscience ; 387: 92-103, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29274353

ABSTRACT

Earlier studies indicate that the central nucleus of the amygdala (CeA) contributes to neuropathic pain. Here we studied whether amygdaloid administration of antioxidants or antagonists of TRPA1 that is among ion channels activated by oxidative stress attenuates nociceptive or affective pain in experimental neuropathy, and whether this effect involves amygdaloid astrocytes or descending serotonergic pathways acting on the spinal 5-HT1A receptor. The experiments were performed in rats with spared nerve injury (SNI). Drugs were administered through a chronic cannula in the CeA or internal capsule (control site), and an intrathecal catheter. Nociception was assessed using monofilaments and affective pain using conditioned place-aversion. Antioxidants or TRPA1 antagonists in the CeA attenuated both nociceptive and affective pain in SNI animals but not in sham controls or in a control injection site. Drugs influencing astroglia (a gap junction decoupler or a D-amino acid oxidase inhibitor) in the CeA had no effect on SNI rats, whereas local anesthesia of the CeA attenuated nociception. Spinally administered 5-HT1A receptor antagonist at a dose that had no effect alone prevented the antinociceptive effect of amygdaloid TRPA1 blockers. The results suggest that injury-induced amygdaloid oxidative stress that drives TRPA1 promotes neuropathic pain behavior. This pronociceptive effect involves suppression of medullospinal serotonergic feedback-inhibition acting on the spinal 5-HT1A receptor. While the CeA is involved in mediating the nerve injury-induced pronociception, it may not be a critical relay for the recruitment of medullospinal feedback-inhibition.


Subject(s)
Amygdala/metabolism , Neuralgia/metabolism , Oxidative Stress , Amygdala/drug effects , Animals , Carbenoxolone/pharmacology , Conditioning, Psychological/drug effects , Cyclic N-Oxides/pharmacology , Internal Capsule/drug effects , Lidocaine/pharmacology , Male , Microinjections , Neuralgia/prevention & control , Oxidative Stress/drug effects , Oximes/pharmacology , Pain Measurement/drug effects , Peripheral Nerve Injuries , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Spin Labels , tert-Butylhydroperoxide/pharmacology
4.
Eur J Pain ; 21(9): 1505-1515, 2017 10.
Article in English | MEDLINE | ID: mdl-28493519

ABSTRACT

BACKGROUND: Repetitive transcranial magnetic stimulation (rTMS) at M1/S1 cortex has been shown to alleviate neuropathic pain. OBJECTIVES: To investigate the possible neurobiological correlates of cortical neurostimulation for the pain relief. METHODS: We studied the effects of M1/S1 rTMS on nociception, brain dopamine D2 and µ-opioid receptors using a randomized, sham-controlled, double-blinded crossover study design and 3D-positron emission tomography (PET). Ten healthy subjects underwent active and sham rTMS treatments to the right M1/S1 cortex with E-field navigated device. Dopamine D2 and µ-receptor availabilities were assessed with PET radiotracers [11 C]raclopride and [11 C]carfentanil after each rTMS treatment. Thermal quantitative sensory testing (QST), contact heat evoked potential (CHEP) and blink reflex (BR) recordings were performed between the PET scans. RESULTS: µ-Opioid receptor availability was lower after active than sham rTMS (P ≤ 0.0001) suggested release of endogenous opioids in the right ventral striatum, medial orbitofrontal, prefrontal and anterior cingulate cortices, and left insula, superior temporal gyrus, dorsolateral prefrontal cortex and precentral gyrus. There were no differences in striatal dopamine D2 receptor availability between active and sham rTMS, consistent with lack of long-lasting measurable dopamine release. Active rTMS potentiated the dopamine-regulated habituation of the BR compared to sham (P = 0.02). Thermal QST and CHEP remained unchanged after active rTMS. CONCLUSIONS: rTMS given to M1/S1 activates the endogenous opioid system in a wide brain network associated with processing of pain and other salient stimuli. Direct enhancement of top-down opioid-mediated inhibition may partly explain the clinical analgesic effects of rTMS. SIGNIFICANCE: Neurobiological correlates of rTMS for the pain relief are unclear. rTMS on M1/S1 with 11 C-carfentanyl-PET activates endogenous opioids. Thermal and heat pain thresholds remain unchanged. rTMS induces top-down opioid-mediated inhibition but not change the sensory discrimination of painful stimuli.


Subject(s)
Cerebral Cortex/metabolism , Opioid Peptides/metabolism , Pain Management , Pain/metabolism , Positron-Emission Tomography , Transcranial Magnetic Stimulation/methods , Adult , Cerebral Cortex/diagnostic imaging , Cross-Over Studies , Female , Humans , Male , Pain/diagnostic imaging , Pain Measurement , Pain Threshold/physiology , Receptors, Dopamine D2/metabolism , Receptors, Opioid, mu/metabolism , Young Adult
5.
J Physiol Pharmacol ; 67(4): 587-594, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27779479

ABSTRACT

Methylglyoxal (MG) is a reactive carbonyl compound generated in diabetes mellitus. MG is an established transient receptor potential ankyrin 1 (TRPA1) channel agonist that contributes to TRPA1-mediated diabetic pain hypersensitivity. Here we studied whether exposure to diabetes and thereby to elevated endogenous MG modulates hypersensitivity induced by intradermal MG. Moreover, since diabetes induces endoplasmic reticulum (ER) stress, we compared the role of TRPA1 in diabetes and ER stress by assessing whether tunicamycin-induced ER stress, without diabetes, produces TRPA1-mediated pain hypersensitivity and by assessing whether ER stress and diabetes have similar modulatory effects on MG-induced hypersensitivity. In vitro patch clamp recording was performed to assess whether tunicamycin is a TRPA1 agonist. Behavioral tests showed that mechanical hypersensitivity induced by MG is reduced in diabetes and ER stress. In healthy controls, hypersensitivity induced by MG was reduced when MG was administered for the second time in the same but not adjacent plantar sites. Hypersensitivity induced by ER stress was reversed by pharmacological blocking of TRPA1. In vitro patch clamp recording indicated that tunicamycin itself (30 µM) is not a TRPA1 agonist. The results indicate that pain hypersensitivity induced by non-diabetic ER stress as well as that induced by diabetes is mediated TRPA1. Reduction of MG-induced hypersensitivity in diabetes or ER stress may, at least partly, be explained by peripheral mechanisms.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hyperalgesia/chemically induced , Pyruvaldehyde/pharmacology , TRPC Cation Channels/agonists , Tunicamycin/pharmacology , Administration, Cutaneous , Animals , Behavior, Animal/drug effects , Diabetes Mellitus, Experimental , HEK293 Cells , Humans , Male , Pain Measurement , Physical Stimulation , Rats , Skin/drug effects , TRPA1 Cation Channel , TRPC Cation Channels/physiology
6.
Neuroscience ; 312: 108-19, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26548413

ABSTRACT

The involvement of the prefrontal cortex in pain processing has been recently addressed. We studied the role of the infralimbic cortex (IL) and group I metabotropic glutamate receptors (mGluRs) in descending modulation of nociception in control and monoarthritic (ARTH) conditions. Nociception was assessed using heat-induced paw withdrawal while drugs were microinjected in the IL of rats. Local anesthesia of the IL or the adjacent prelimbic cortex (PL) facilitated nociception, indicating that IL and PL are tonically promoting spinal antinociception. Phasic activation with glutamate (GLU) revealed opposing roles of the PL and IL; GLU in the PL had a fast antinociceptive action, while in the IL it had a slow onset pronociceptive action. IL administration of a local anesthetic or GLU produced identical results in ARTH and control animals. An mGluR5 agonist in the IL induced a pronociceptive effect in both groups, while mGluR5 antagonists had no effect in controls but induced antinociception in ARTH rats. Activation of the IL mGluR1 (through co-administration of mGluR1/5 agonist and mGluR5 antagonist) did not alter nociception in controls but induced antinociception in ARTH animals. IL administration of an mGluR1 antagonist failed to alter nociception in either experimental group. Finally, mGluR5 but not mGluR1 antagonists blocked the pronociceptive action of GLU in both groups. The results indicate that IL contributes to descending modulation of nociception. mGluR5 in the IL enhance nociception in healthy control and monoarthritic animals, an effect that is tonic in ARTH. Moreover, activation of IL mGluR1s attenuates nociception following the development of monoarthritis.


Subject(s)
Arthritis/physiopathology , Glutamic Acid/physiology , Hyperalgesia/physiopathology , Nociception/physiology , Prefrontal Cortex/physiology , Receptor, Metabotropic Glutamate 5/physiology , Animals , Disease Models, Animal , Male , Nociception/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
7.
Behav Brain Res ; 282: 14-24, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25557801

ABSTRACT

Amygdala is involved in processing of primary emotions and particularly its central nucleus (CeA) also in pain control. Here we studied mechanisms mediating the descending control of mechanical hypersensitivity by the CeA in rats with a peripheral neuropathy in the left hind limb. For drug administrations, the animals had a guide cannula in the right CeA and an intrathecal catheter or another guide cannula in the medullary raphe. Hypersensitivity was tested with monofilaments. Glutamate administration in the CeA produced a bidirectional effect on hypersensitivity that varied from an increase at a low-dose (9µg) to a reduction at high doses (30-100µg). The increase but not the reduction of hypersensitivity was prevented by blocking the amygdaloid NMDA receptor with a dose of MK-801 that alone had no effects. The glutamate-induced increase in hypersensitivity was reversed by blocking the spinal 5-HT3 receptor with ondansetron, whereas the reduction in hypersensitivity was reversed by blocking the spinal 5-HT1A receptor with WAY-100635. Both the increase and decrease of hypersensitivity induced by amygdaloid glutamate treatment were reversed by medullary administration of a 5-HT1A agonist, 8-OH-DPAT, that presumably produced autoinhibition of serotonergic cell bodies in the medullary raphe. The results indicate that depending on the dose, glutamate in the CeA has a descending facilitatory or inhibitory effect on neuropathic pain hypersensitivity. Serotoninergic raphe neurons are involved in mediating both of these effects. Spinally, the 5-HT3 receptor contributes to the increase and the 5-HT1A receptor to the decrease of neuropathic hypersensitivity induced by amygdaloid glutamate.


Subject(s)
Amygdala/metabolism , Neuralgia/metabolism , Peripheral Nervous System Diseases/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Spinal Cord/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Amygdala/drug effects , Animals , Dizocilpine Maleate/pharmacology , Glutamic Acid/pharmacology , Male , Ondansetron/pharmacology , Pain Measurement/methods , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin 5-HT3 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Spinal Cord/drug effects
8.
Behav Brain Res ; 265: 12-21, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24518202

ABSTRACT

Affective disorders are common comorbidities of chronic inflammatory pain that are often overlooked in primary care. As the impact of inflammatory pain upon mood-like disorders in animal models is not well known, our objective was to assess whether prolonged experimental monoarthritis (ARTH) induced the development of anxiety and depressive-like behaviours in rodents and if amitriptyline, an antidepressant commonly used in the treatment of chronic pain, could reverse both nociceptive and mood-like impairments. Experimental ARTH was induced through an injection of kaolin/carrageenan into the right knee joint with control (SHAM) animals injected with saline. Four weeks after induction, ARTH animals displayed mechanical hyperalgesia and a depressive-like phenotype as they showed a significant increase in immobility and a decrease in the latency to immobility in the forced-swimming test at the expense of the time spent climbing/swimming. ARTH animals also displayed a decreased sucrose preference, an index of anhedonia and anxiety-like behaviour as time spent exploring the open arms of the elevated-plus-maze was decreased when compared to controls. The anxiety-like phenotype was also supported by an increase in the number of fecal boli left in the open field. In ARTH animals, the administration of amitriptyline decreased mechanical hyperalgesia and increased sucrose preference and the time spent climbing, although it had a deleterious effect in the performance of control animals. Our data show that this model of ARTH can be useful for the study of chronic pain-mood disorders comorbidities and that amitriptyline is able to partly reverse the associated nociceptive and emotional impairments.


Subject(s)
Amitriptyline/pharmacology , Amitriptyline/therapeutic use , Analgesics, Non-Narcotic/therapeutic use , Arthritis/complications , Hyperalgesia/drug therapy , Mood Disorders/drug therapy , Mood Disorders/etiology , Animals , Arthritis/chemically induced , Carrageenan/pharmacology , Disease Models, Animal , Exploratory Behavior/drug effects , Food Preferences/drug effects , Hyperalgesia/etiology , Kaolin/pharmacology , Male , Maze Learning/drug effects , Motor Activity/drug effects , Pain Threshold/drug effects , Physical Stimulation/adverse effects , Rats , Rats, Wistar , Swimming/psychology
9.
Neuroscience ; 253: 426-34, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24036376

ABSTRACT

The neurobiological mechanisms of spinal cord stimulation (SCS) when applied for neuropathic pain are still incompletely known. Previous research indicates that brainstem circuitry is pivotal for the SCS effect. The present study aims at exploring the possible contribution to the SCS effects of the pain controlling system emanating from the locus coeruleus (LC) in the brain stem. Experiments were performed on the rat-spared nerve injury pain model. After evaluation of the attenuation of mechanical hypersensitivity induced by SCS, the effects of SCS on neuronal activity in the LC and on the noradrenaline (NA) content in the dorsal spinal cord were analyzed. SCS produced a significant increase in the discharge rate of LC neurons only in rats behaviorally responding to SCS as compared to non-responding and control animals. The NA content in the dorsal quadrant of the spinal cord ipsilateral to the nerve injury was analyzed using enzyme-linked immunosorbent assay in responding, non-responding and intact control rats both immediately following SCS and without SCS. No differences were found between these groups. In awake animals, lidocaine silencing of the ipsilateral LC or blocking of spinal noradrenergic system by intrathecal administration of α1,2 adrenoceptor antagonists failed to influence the antihypersensitivity effect of SCS. The present results indicate that the SCS-induced control of hypersensitivity in an experimental animal model of peripheral neuropathic pain may not be explained by the activation of direct spinal projections of noradrenergic LC neurons, while supraspinal projections of LC neurons still may play a role in the SCS effect.


Subject(s)
Electric Stimulation , Locus Coeruleus/pathology , Neurons/physiology , Sciatica/therapy , Spinal Cord/physiology , Action Potentials/drug effects , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Anesthetics, Local/pharmacology , Animals , Disease Models, Animal , Hyperalgesia/physiopathology , Hyperalgesia/therapy , Imidazoles/pharmacology , Lidocaine/pharmacology , Locus Coeruleus/drug effects , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Pain Threshold/physiology , Physical Stimulation/adverse effects , Prazosin/pharmacology , Rats , Rats, Wistar
10.
J Physiol Pharmacol ; 64(3): 331-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23959730

ABSTRACT

We characterized pain behavior and cutaneous blood flow response induced by activation of the spinal transient receptor potential ankyrin 1 (TRPA1) channel using intrathecal drug administrations in the rat. Additionally, we assessed whether the pronociceptive actions induced by intrathecally administered dynorphin A, cholecystokinin or prostaglandin F(2α) are mediated by the spinal TRPA1 channel. Cinnamaldehyde, a TRPA1 agonist, produced a dose-related (3-10 µg) cutaneous blood flow increase and mechanical hypersensitivity effect. These effects at the currently used doses were of short duration and attenuated, although not completely, by pretreatment with A-967079, a TRPA1 antagonist. The cinnamaldehyde-induced hypersensitivity was also reduced by pretreatment with minocycline (an inhibitor of microglial activation), but not by carbenoxolone (a gap junction decoupler). In vitro study, however, indicated that minocycline only poorly blocks the TRPA1 channel. The mechanical hypersensitivity effect induced by dynorphin A, but not that by cholecystokinin or prostaglandin F(2α), was attenuated by a TRPA1 antagonist Chembridge-5861528 as well as A-967079. The cinnamaldehyde-induced cutaneous blood flow increase was not suppressed by MK-801, an NMDA receptor antagonist, or bicuculline, a GABA(A) receptor antagonist. The results indicate that spinal TRPA1 channels promote mechanical pain hypersensitivity and due to antidromic activation of nociceptive nerve fibers increase cutaneous blood flow. The attenuation of the cinnamaldehyde-induced hypersensitivity effect by minocycline may be explained by action other than block of the TRPA1 channel. Moreover, the spinal TRPA1 channel is involved in mediating the pronociceptive action of dynorphin A, but not that of the spinal cholecystokinin or prostaglandin F(2α).


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Back Pain/drug therapy , Dynorphins/antagonists & inhibitors , Hyperalgesia/drug therapy , Posterior Horn Cells/drug effects , Skin/drug effects , TRPC Cation Channels/antagonists & inhibitors , Acrolein/administration & dosage , Acrolein/adverse effects , Acrolein/analogs & derivatives , Acrolein/antagonists & inhibitors , Analgesics, Non-Narcotic/administration & dosage , Animals , Back Pain/etiology , Back Pain/metabolism , Behavior, Animal/drug effects , Cholecystokinin/administration & dosage , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/metabolism , Dinoprost/administration & dosage , Dinoprost/antagonists & inhibitors , Dinoprost/metabolism , Dose-Response Relationship, Drug , Dynorphins/administration & dosage , Dynorphins/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Injections, Spinal , Male , Minocycline/administration & dosage , Minocycline/therapeutic use , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Oximes/administration & dosage , Oximes/therapeutic use , Physical Stimulation/adverse effects , Posterior Horn Cells/metabolism , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Skin/blood supply , TRPA1 Cation Channel , TRPC Cation Channels/agonists , TRPC Cation Channels/metabolism
11.
Neuroscience ; 247: 134-44, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23711584

ABSTRACT

The neurobiological mechanisms underlying the suppression of neuropathic pain by spinal cord stimulation (SCS) are still incompletely known. The present study aims at exploring whether the descending pain control system in the rostroventromedial medulla (RVM) exerts a role in the attenuation of neuropathic pain by SCS. Experiments were performed in the rat spared nerve injury (SNI) pain model. The effects of SCS on neuronal activity of pronociceptive ON-like, antinociceptive OFF-like, and neutral cells, including 5-HT-like cells, in the RVM were analyzed in SCS responding and SCS non-responding SNI animals as well as in naïve controls. Decreased spontaneous activities in OFF-like cells and increased spontaneous activities in ON-like cells were observed in SNI animals, whereas the spontaneous activities of 5-HT-like and neutral cells were unchanged. SCS produced a prominent increase in the discharge of OFF- and 5-HT-like cells in SCS responding, but not in non-responding SNI animals or controls. Discharge rates of ON-like and neutral cell were not affected by SCS. In awake SNI animals, microinjection of a GABAA receptor agonist, muscimol, into the RVM significantly attenuated the antihypersensitivity effect induced by SCS while a non-selective opioid receptor antagonist, naltrexone, was ineffective. It is concluded that SCS may shift the reciprocal inhibitory and facilitatory pain modulation balance controlled by the RVM in favor of inhibition. This increase in the descending antinociceptive effect operates in concert with segmental spinal mechanisms in producing pain relief.


Subject(s)
Medulla Oblongata/physiology , Neuralgia/therapy , Pain Management/methods , Pain Measurement/methods , Spinal Cord Stimulation/methods , Animals , Male , Neural Pathways/physiology , Neuralgia/physiopathology , Rats , Rats, Wistar , Treatment Outcome
12.
Genes Brain Behav ; 11(5): 513-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22435671

ABSTRACT

Fragile X syndrome (FXS) is a common cause of inherited intellectual disability and a well-characterized form of autism spectrum disorder. As brain-derived neurotrophic factor (BDNF) is implicated in the pathophysiology of FXS we examined the effects of reduced BDNF expression on the behavioral phenotype of an animal model of FXS, Fmr1 knockout (KO) mice, crossed with mice carrying a deletion of one copy of the Bdnf gene (Bdnf(+/-)). Fmr1 KO mice showed age-dependent alterations in hippocampal BDNF expression that declined after the age of 4 months compared to wild-type controls. Mild deficits in water maze learning in Bdnf(+/-) and Fmr1 KO mice were exaggerated and contextual fear learning significantly impaired in double transgenics. Reduced BDNF expression did not alter basal nociceptive responses or central hypersensitivity in Fmr1 KO mice. Paradoxically, the locomotor hyperactivity and deficits in sensorimotor learning and startle responses characteristic of Fmr1 KO mice were ameliorated by reducing BNDF, suggesting changes in simultaneously and in parallel working hippocampus-dependent and striatum-dependent systems. Furthermore, the obesity normally seen in Bdnf(+/-) mice was eliminated by the absence of fragile X mental retardation protein 1 (FMRP). Reduced BDNF decreased the survival of newborn cells in the ventral part of the hippocampus both in the presence and absence of FMRP. Since a short neurite phenotype characteristic of newborn cells lacking FMRP was not found in cells derived from double mutant mice, changes in neuronal maturation likely contributed to the behavioral phenotype. Our results show that the absence of FMRP modifies the diverse effects of BDNF on the FXS phenotype.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cognition Disorders/genetics , Fragile X Mental Retardation Protein/genetics , Gait Disorders, Neurologic/genetics , Hyperkinesis/genetics , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cognition Disorders/metabolism , Conditioning, Psychological/physiology , Fear/physiology , Fragile X Mental Retardation Protein/metabolism , Gait Disorders, Neurologic/metabolism , Hippocampus/metabolism , Hyperkinesis/metabolism , Maze Learning/physiology , Mice , Mice, Knockout , Motor Activity/genetics , Neural Stem Cells , Neurons/metabolism , Reflex, Startle/genetics
13.
Br J Pharmacol ; 161(7): 1553-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20726980

ABSTRACT

BACKGROUND AND PURPOSE: Catechol-O-methyltransferase (COMT) inhibitors are used in Parkinson's disease in which pain is an important symptom. COMT polymorphisms modulate pain and opioid analgesia in humans. In rats, COMT inhibitors have been shown to be pro-nociceptive in acute pain models, but also to attenuate allodynia and hyperalgesia in a model of diabetic neuropathy. Here, we have assessed the effects of acute and repeated administrations of COMT inhibitors on mechanical, thermal and carrageenan-induced nociception in male mice. EXPERIMENTAL APPROACH: We used single and repeated administration of a peripherally restricted, short-acting (nitecapone) and also a centrally acting (3,5-dinitrocatechol, OR-486) COMT inhibitor. We also tested CGP 28014, an indirect inhibitor of COMT enzyme. Effects of OR-486 on thermal nociception were also studied in COMT deficient mice. Effects on spinal pathways were assessed in rats given intrathecal nitecapone. KEY RESULTS: After single administration, both nitecapone and OR-486 reduced mechanical nociceptive thresholds and thermal nociceptive latencies (hot plate test) at 2 and 3 h, regardless of their brain penetration. These effects were still present after chronic treatment with COMT inhibitors for 5 days. Intraplantar injection of carrageenan reduced nociceptive latencies and both COMT inhibitors potentiated this reduction without modifying inflammation. CGP 28014 shortened paw flick latencies. OR-486 did not modify hot plate times in Comt gene deficient mice. Intrathecal nitecapone modified neither thermal nor mechanical nociception. CONCLUSIONS AND IMPLICATIONS: Pro-nociceptive effects of COMT inhibitors were confirmed. The pro-nociceptive effects were primarily mediated via mechanisms acting outside the brain and spinal cord. COMT protein was required for these actions.


Subject(s)
Catechol O-Methyltransferase Inhibitors , Catechol O-Methyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Nociceptors/physiology , Pain Threshold/drug effects , Pain/chemically induced , Animals , Behavior, Animal/drug effects , Catechol O-Methyltransferase/genetics , Catechols/administration & dosage , Catechols/pharmacology , Enzyme Inhibitors/administration & dosage , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/physiopathology , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain/physiopathology , Pain Measurement , Pentanones/administration & dosage , Pentanones/pharmacology , Rats , Rats, Wistar , Time Factors
14.
Neuroscience ; 167(4): 1175-82, 2010 Jun 02.
Article in English | MEDLINE | ID: mdl-20298759

ABSTRACT

Memantine is a low-affinity NMDA receptor antagonist that is used in the treatment of Alzheimer's disease to alleviate the cognitive symptoms of the disease. In humans, memantine has been shown to facilitate auditory change detection as reflected in the mismatch negativity (MMN) response recorded in the frontal cortex. In the present study we investigated the effects of memantine on the auditory MMN-like responses recorded in anesthetized rats. Saline, a low (3 mg/kg) or a high (10 mg/kg) dose of memantine was i.p. injected into the animals. Auditory MMN-like responses were recorded during the presentation of a repeated tone of one frequency (standard, P=0.956) that was rarely replaced by a tone of another frequency (deviant, P=0.044). The low dose of memantine did not observably affect the amplitude of the auditory MMN-like response, but it prolonged the duration of the response relative to saline. The high dose of memantine, in contrast, blocked the generation of the auditory MMN-like response. The findings suggest that memantine may, with appropriate doses, facilitate already this early stage of auditory processing.


Subject(s)
Evoked Potentials, Auditory/drug effects , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Acoustic Stimulation , Anesthesia , Animals , Auditory Cortex/drug effects , Auditory Cortex/physiology , Contingent Negative Variation/drug effects , Dose-Response Relationship, Drug , Male , Rats
15.
Neuroscience ; 146(4): 1785-94, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17445989

ABSTRACT

Noradrenergic locus coeruleus (LC) is involved in pain regulation. We studied whether response properties of LC neurons or coeruleospinal antinociception are changed 10-14 days following development of experimental neuropathy. Experiments were performed in spinal nerve-ligated, sham-operated and unoperated male rats under sodium pentobarbital anesthesia. Recordings of LC neurons indicated that responses evoked by noxious somatic stimulation were enhanced in nerve-injured animals, while the effects of nerve injury on spontaneous activity or the response to noxious visceral stimulation were not significant. Microinjection of glutamate into the central nucleus of the amygdala produced a dose-related inhibition of the discharge rate of LC neurons in nerve-injured animals but no significant effect on discharge rates in control groups. Assessment of the heat-induced hind limb withdrawal latency indicated that spinal antinociception induced by electrical stimulation of the LC was significantly weaker in nerve-injured than control animals. The results indicate that peripheral neuropathy induces bidirectional changes in coeruleospinal inhibition of pain. Increased responses of LC neurons to noxious somatic stimulation are likely to promote feedback inhibition of neuropathic hypersensitivity, while the enhanced inhibition of the LC from the amygdala is likely to suppress noradrenergic pain inhibition and promote neuropathic pain. It is proposed that the decreased spinal antinociception induced by direct stimulation of the LC may be explained by pronociceptive changes in the non-noradrenergic systems previously described in peripheral neuropathy. Furthermore, we propose the hypothesis that emotions processed by the amygdala enhance pain due to increased inhibition of the LC in peripheral neuropathy.


Subject(s)
Hyperalgesia/etiology , Locus Coeruleus/pathology , Neurons/physiology , Peripheral Nervous System Diseases/complications , Action Potentials/physiology , Animals , Behavior, Animal , Dose-Response Relationship, Radiation , Efferent Pathways/physiopathology , Electric Stimulation/methods , Functional Laterality/physiology , Glutamic Acid/pharmacology , Locus Coeruleus/radiation effects , Male , Neurons/drug effects , Neurons/radiation effects , Pain Measurement/methods , Peripheral Nervous System Diseases/pathology , Rats , Rats, Wistar , Reaction Time/physiology , Reaction Time/radiation effects
16.
Neuroscience ; 134(3): 1023-32, 2005.
Article in English | MEDLINE | ID: mdl-16039797

ABSTRACT

The mammalian RFamide-related peptide RFRP1 was found to signal through the neuropeptide FF 2 receptor expressed in Xenopus oocytes. The peptide induced a dose-dependent outward current, which was dependent on the simultaneous expression of GIRK1 and GIRK4 potassium channels. In neuropathic rats, RFRP1 administered intrathecally induced tactile antiallodynia and thermal antinociception, whereas in the solitary tract nucleus it produced only mechanical antihyperalgesia. Expression of the RFamide-related peptide mRNA in the rat CNS was distinctly different from that of neuropeptide FF. Most notably, the gene was not expressed in the hindbrain or spinal cord at detectable levels. However, there was a prominent group of RFamide-related peptide mRNA-expressing neurons in the central hypothalamus, in the area in and between the dorsomedial and ventromedial nuclei. The results suggest that RFamide-related peptides are potentially involved in pain regulation through a hypothalamo-medullary projection system, and possibly via action on neuropeptide FF 2 receptors. In neuropathic animals, the pain suppressive effect of RFamide-related peptide varies depending on the submodality of noxious test stimulation and the site of RFamide-related peptide administration.


Subject(s)
Neuropeptides/administration & dosage , Pain/drug therapy , Receptors, Neuropeptide/physiology , Signal Transduction/physiology , Animals , Cloning, Molecular/methods , Dose-Response Relationship, Drug , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , In Situ Hybridization/methods , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections/methods , Neuropeptides/pharmacology , Pain/physiopathology , Pain Measurement/methods , Pain Threshold/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reaction Time/drug effects , Solitary Nucleus/drug effects , Solitary Nucleus/physiopathology , Spinal Cord/drug effects , Spinal Cord/physiopathology , Xenopus laevis
17.
Neuroscience ; 114(3): 655-68, 2002.
Article in English | MEDLINE | ID: mdl-12220567

ABSTRACT

We studied the effect of chemical sympathectomy by 6-hydroxydopamine (6-OHDA) on pain behavior and alpha(2)-adrenergic antinociception in rats with a spinal nerve ligation-induced neuropathy. For assessment of alpha(2)-adrenergic antinociception, the rats were treated systemically with two alpha(2)-adrenoceptor agonists, one of which only poorly (MPV-2426) and the other very well (dexmedetomidine) penetrates the blood-brain barrier. Moreover, the effect of MPV-2426 on spontaneous activity of dorsal root nerve fibers proximal to the nerve injury was determined. Systemic treatment with 6-OHDA produced a marked decrease in immunocytochemical labeling of sympathetic nerve fibers in the skin but it produced no marked change in basal pain sensitivity to mechanical stimulation either in neuropathic or sham-operated animals. Systemic administration of MPV-2426 and dexmedetomidine produced a dose-dependent tactile antiallodynic effect in neuropathic animals. Intraplantar injection of MPV-2426 had an identical antiallodynic effect independent of whether it was injected into the neuropathic or contralateral hindpaw. In a test of mechanical nociception and hyperalgesia, dexmedetomidine markedly attenuated pain responses in all experimental groups, whereas MPV-2426 had a weak but significant pain attenuating effect only in neuropathic animals. In the tail flick test, both alpha(2)-adrenoceptor agonists had a significant antinociceptive effect. The pain attenuating effect of MPV-2426 was enhanced by pretreatment with 6-OHDA, except in a test of tactile allodynia. MPV-2426-induced modulation of spontaneous activity was not a general property of dorsal root fibers proximal to the injury. The results indicate that a chemical destruction of sympathetic postganglionic nerve fibers innervating the skin does not markedly influence cutaneous pain sensitivity nor is it critical for the alpha(2)-adrenoceptor agonist-induced attenuation of pain behavior in neuropathic or non-neuropathic animals. Chemical sympathectomy, independent of neuropathy, enhanced the pain attenuating effect by MPV-2426, probably due to a peripheral action, whereas in non-sympathectomized control and neuropathic animals peripheral mechanisms have only a minor, if any, role in the alpha(2)-adrenoceptor agonist-induced antinociception.


Subject(s)
Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Analgesics/pharmacology , Pain Measurement/drug effects , Sympathectomy, Chemical , Animals , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Imidazoles/pharmacology , Indans/pharmacology , Ligation , Male , Pain Measurement/methods , Pain Measurement/statistics & numerical data , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/physiology , Spinal Nerves/injuries , Sympathectomy, Chemical/methods , Sympathectomy, Chemical/statistics & numerical data , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Fibers, Postganglionic/physiology
18.
Neuroscience ; 113(2): 339-49, 2002.
Article in English | MEDLINE | ID: mdl-12127091

ABSTRACT

The purpose of the present study was to investigate the role of mu-opioid receptor in inflammatory hyperalgesia in intact and in spinalized animals and the interaction between mu-opioid and alpha2-adrenergic receptor in acute pain and inflammatory hyperalgesia. Behavioral responses to mechanical and heat stimuli were studied in mu-opioid receptor knockout mice and wildtype control mice. Thermal nociception was evaluated by measuring paw withdrawal latencies to radiant heat applied to the hindpaws. Mechanical nociception was measured by von Frey monofilament applications to the hindpaws. Intraplantar carrageenan-induced (1 mg/40 microl) mechanical and heat hyperalgesia were compared in micro-opioid knockout and wildtype mice. The effect of systemically administered alpha2-adrenergic receptor agonist dexmedetomidine (1-10 microg/kg) was evaluated on mechanical and thermal withdrawal responses under normal and inflammatory state in knockout and wildtype mice. The role of micro-opioid receptor in descending modulation of nociception was studied by assessing mechanical and heat withdrawal responses before and after mid-thoracic spinalization. Withdrawal responses to radiant heat and von Frey monofilaments were similar in mu-opioid knockout and wildtype mice before and after the carrageenan induced hindpaw inflammation. Also, antinociceptive effects of dexmedetomidine in thermal and mechanical nociceptive tests were similar before carrageenan induced hindpaw inflammation. However, the potency of dexmedetomidine was significantly reduced in carrageenan-induced mechanical hyperalgesia in mu-opioid knockout mice compared to the wildtype control mice. Thermal and mechanical withdrawal responses were similar between mu-opioid knockout and wildtype mice before and after mid-thoracic spinalization. Our observations indicate that the mu-opioid receptors do not play an important role in alpha2-adrenergic receptor agonist-mediated acute antinociception. In addition, micro-opioid receptors are not tonically involved in the modulation of inflammation-induced mechanical and thermal hyperalgesia, and the supraspinal control of spinal reflexes. However, in the presence of inflammation, mu-opioid receptors play an important role in the antihyperalgesic actions of an alpha2-adrenergic receptor agonist.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/physiopathology , Inflammation/complications , Receptors, Adrenergic, alpha/physiology , Receptors, Opioid, mu/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Carrageenan , Decerebrate State , Dexmedetomidine/pharmacology , Foot , Hindlimb , Hot Temperature , Imidazoles/pharmacology , Inflammation/chemically induced , Mice , Mice, Knockout/genetics , Nociceptors/drug effects , Physical Stimulation , Receptors, Opioid, mu/genetics , Reference Values
19.
Eur J Pharmacol ; 429(1-3): 139-45, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11698036

ABSTRACT

We studied the contribution of peripheral opioid receptors to the morphine-induced antinociception in rats with a spinal nerve ligation-induced neuropathy. Intraplantar (i.pl.) injection of morphine produced a stronger suppression of nociceptive reflex responses of the neuropathic limb following ipsilateral, than contralateral, administration, whereas the morphine-induced effect on the control limb was independent of the injection side. Antinociception induced by systemically administered morphine was significantly attenuated by i.pl. injection of a peripherally acting opioid receptor antagonist in neuropathic but not in sham-operated rats. Following chemical sympathectomy with 6-hydroxydopamine, antinociception was achieved at a lower dose ipsilaterally, than contralaterally, following i.pl. administration of morphine, and the morphine-induced antinociception was attenuated by a peripherally acting opioid receptor antagonist. These results indicate that peripheral opioid receptors may contribute to the morphine-induced antinociception in the spinal nerve ligation-induced model of neuropathy. Sympathectomy of the neuropathic limb may underlie, at least partly, the increased peripheral efficacy of morphine in neuropathy.


Subject(s)
Analgesics, Opioid/administration & dosage , Mononeuropathies/drug therapy , Morphine/administration & dosage , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Fibers, Postganglionic/physiology , Animals , Hindlimb , Injections, Subcutaneous , Ligation , Male , Mononeuropathies/chemically induced , Oxidopamine , Pain Measurement/drug effects , Rats , Rats, Wistar , Receptors, Opioid/physiology , Spinal Nerves/drug effects , Spinal Nerves/physiology , Sympatholytics
20.
Neuroscience ; 105(2): 457-68, 2001.
Article in English | MEDLINE | ID: mdl-11672611

ABSTRACT

We determined whether chronic neuropathy changes response properties of neurons in the rostroventromedial medulla of rats, and whether (d-Tyr)L(Me-Phe)QPQRF-amide, a neuropeptide FF analogue, in the periaqueductal gray produces changes in responses of rostroventromedial medullary neurons that might underlie its antiallodynic effect described earlier. Single unit recordings of medullary neurons were performed in lightly anesthetized neuropathic and control animals. Spontaneous activity and the responses to noxious thermal and mechanical stimulation of the hind paw were determined with and without administration of (d-Tyr)L(Me-Phe)QPQRF-amide. The neurons were classified into three groups: ON-neurons increased, OFF-neurons decreased, and NEUTRAL-neurons did not change their discharge rate prior to a limb withdrawal induced by noxious stimulation of the skin. Spontaneous activity and heat-evoked responses of ON-neurons were not different between neuropathic and control animals, whereas their mechanically evoked responses were reduced in neuropathy. Response properties of OFF-neurons were not different between neuropathic and control animals. Spontaneous activity of NEUTRAL-neurons was not different between neuropathic and control animals. (d-Tyr)L(Me-Phe)QPQRF-amide in the periaqueductal gray had no significant effect on evoked responses or spontaneous activity of ON- or OFF-neurons, independent of the experimental group. However, (d-Tyr)L(Me-Phe)QPQRF-amide produced a significant attenuation of spontaneous activity of NEUTRAL-neurons in neuropathic animals. In a behavioral study performed in unanesthetized animals it was found that intrathecal administration of methysergide, a serotonin antagonist, selectively attenuated neuropathic symptoms. Also, light pentobarbitone anesthesia markedly attenuated, but did not abolish, behaviorally determined neuropathic symptoms. From these results we suggest that NEUTRAL-neurons of the rostroventromedial medulla may have a role in neuropathy and they may be involved in attenuation of mechanical hypersensitivity by (d-Tyr)L(Me-Phe)QPQRF-amide in the periaqueductal gray. It is proposed that in neuropathy the synaptic effects of descending impulses from medullary NEUTRAL-neurons on their axonal targets in the spinal cord are changed so that this contributes to mechanical hypersensitivity, due to mechanisms that are at least partly serotoninergic.


Subject(s)
Action Potentials/physiology , Contrast Media/pharmacology , Medulla Oblongata/metabolism , Neuralgia/metabolism , Oligopeptides/pharmacology , Raphe Nuclei/metabolism , Reticular Formation/metabolism , Action Potentials/drug effects , Adjuvants, Anesthesia/pharmacology , Animals , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Ligation , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Methysergide/pharmacology , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuralgia/pathology , Neuralgia/physiopathology , Oligopeptides/metabolism , Pain Measurement/drug effects , Pentobarbital/pharmacology , Periaqueductal Gray/cytology , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Physical Stimulation , Raphe Nuclei/cytology , Raphe Nuclei/drug effects , Rats , Rats, Wistar , Reaction Time/drug effects , Reaction Time/physiology , Reticular Formation/cytology , Reticular Formation/drug effects , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Spinal Nerves/injuries , Spinal Nerves/physiopathology , Spinal Nerves/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...