Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neuroimage ; 64: 341-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22982372

ABSTRACT

The earliest stages of osteoarthritis are characterized by peripheral pathology; however, during disease progression chronic pain emerges-a major symptom of osteoarthritis linked to neuroplasticity. Recent clinical imaging studies involving chronic pain patients, including osteoarthritis patients, have demonstrated that functional properties of the brain are altered, and these functional changes are correlated with subjective behavioral pain measures. Currently, preclinical osteoarthritis studies have not assessed if functional properties of supraspinal pain circuitry are altered, and if these functional properties can be modulated by pharmacological therapy either by direct or indirect action on brain systems. In the current study, functional connectivity was first assessed in order to characterize the functional neuroplasticity occurring in the rodent medial meniscus tear (MMT) model of osteoarthritis-a surgical model of osteoarthritis possessing peripheral joint trauma and a hypersensitive pain state. In addition to knee joint trauma at week 3 post-MMT surgery, we observed that supraspinal networks have increased functional connectivity relative to sham animals. Importantly, we observed that early and sustained treatment with a novel, peripherally acting broad-spectrum matrix metalloproteinase (MMP) inhibitor (MMPi) significantly attenuates knee joint trauma (cartilage degradation) as well as supraspinal functional connectivity increases in MMT animals. At week 5 post-MMT surgery, the acute pharmacodynamic effects of celecoxib (selective cyclooxygenase-2 inhibitor) on brain function were evaluated using pharmacological magnetic resonance imaging (phMRI) and functional connectivity analysis. Celecoxib was chosen as a comparator, given its clinical efficacy for alleviating pain in osteoarthritis patients and its peripheral and central pharmacological action. Relative to the vehicle condition, acute celecoxib treatment in MMT animals yielded decreased phMRI infusion responses and decreased functional connectivity, the latter observation being similar to what was detected following chronic MMPi treatment. These findings demonstrate that an assessment of brain function may provide an objective means by which to further evaluate the pathology of an osteoarthritis state as well as measure the pharmacodynamic effects of therapies with peripheral or peripheral and central pharmacological action.


Subject(s)
Action Potentials/drug effects , Brain/physiopathology , Disease Models, Animal , Nerve Net/physiopathology , Osteoarthritis/physiopathology , Pain/physiopathology , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Animals , Brain/drug effects , Celecoxib , Humans , Male , Nerve Net/drug effects , Osteoarthritis/complications , Osteoarthritis/drug therapy , Pain/etiology , Pain/prevention & control , Pain Measurement/drug effects , Rats , Rats, Inbred Lew
2.
Bioorg Med Chem Lett ; 22(7): 2604-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22370265

ABSTRACT

SAR studies on a series of thiophene amide derivatives provided CB(2) receptor agonists. The activity of the compounds was characterized by radioligand binding determination, multiple functional assays, ADME, and pharmacokinetic studies. A representative compound with selectivity for CB(2) over CB(1) effectively produced analgesia in behavioral models of neuropathic, inflammatory, and postsurgical pain. Control experiments using a CB(2) antagonist demonstrated the efficacy in the pain models resulted from CB(2) agonism.


Subject(s)
Amides/chemical synthesis , Analgesics/chemical synthesis , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Receptor, Cannabinoid, CB2/agonists , Thiophenes/chemical synthesis , Amides/pharmacokinetics , Amides/pharmacology , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Biological Availability , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hyperalgesia/metabolism , Neuralgia/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Structure-Activity Relationship , Thiophenes/pharmacokinetics , Thiophenes/pharmacology
3.
PLoS One ; 6(11): e27839, 2011.
Article in English | MEDLINE | ID: mdl-22125628

ABSTRACT

During wakefulness and in absence of performing tasks or sensory processing, the default-mode network (DMN), an intrinsic central nervous system (CNS) network, is in an active state. Non-human primate and human CNS imaging studies have identified the DMN in these two species. Clinical imaging studies have shown that the pattern of activity within the DMN is often modulated in various disease states (e.g., Alzheimer's, schizophrenia or chronic pain). However, whether the DMN exists in awake rodents has not been characterized. The current data provides evidence that awake rodents also possess 'DMN-like' functional connectivity, but only subsequent to habituation to what is initially a novel magnetic resonance imaging (MRI) environment as well as physical restraint. Specifically, the habituation process spanned across four separate scanning sessions (Day 2, 4, 6 and 8). At Day 8, significant (p<0.05) functional connectivity was observed amongst structures such as the anterior cingulate (seed region), retrosplenial, parietal, and hippocampal cortices. Prior to habituation (Day 2), functional connectivity was only detected (p<0.05) amongst CNS structures known to mediate anxiety (i.e., anterior cingulate (seed region), posterior hypothalamic area, amygdala and parabracial nucleus). In relating functional connectivity between cingulate-default-mode and cingulate-anxiety structures across Days 2-8, a significant inverse relationship (r = -0.65, p = 0.0004) was observed between these two functional interactions such that increased cingulate-DMN connectivity corresponded to decreased cingulate anxiety network connectivity. This investigation demonstrates that the cingulate is an important component of both the rodent DMN-like and anxiety networks.


Subject(s)
Brain/physiology , Nerve Net/physiology , Wakefulness/physiology , Animals , Anxiety/physiopathology , Brain/anatomy & histology , Brain Mapping , Gyrus Cinguli/anatomy & histology , Gyrus Cinguli/physiology , Hypothalamus/anatomy & histology , Hypothalamus/physiology , Magnetic Resonance Imaging/methods , Male , Models, Neurological , Nerve Net/anatomy & histology , Parietal Lobe/anatomy & histology , Parietal Lobe/physiology , Prefrontal Cortex/anatomy & histology , Prefrontal Cortex/physiology , Rats , Rats, Long-Evans , Respiratory Rate/physiology
4.
Biochem Pharmacol ; 82(8): 967-76, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21620806

ABSTRACT

Positive modulation of the neuronal nicotinic acetylcholine receptor (nAChR) α4ß2 subtype by selective positive allosteric modulator NS-9283 has shown to potentiate the nAChR agonist ABT-594-induced anti-allodynic activity in preclinical neuropathic pain. To determine whether this benefit can be extended beyond neuropathic pain, the present study examined the analgesic activity and adverse effect profile of co-administered NS-9283 and ABT-594 in a variety of preclinical models in rats. The effect of the combined therapy on drug-induced brain activities was also determined using pharmacological magnetic resonance imaging. In carrageenan-induced thermal hyperalgesia, co-administration of NS-9283 (3.5 µmol/kg, i.p.) induced a 6-fold leftward shift of the dose-response of ABT-594 (ED(50)=26 vs. 160 nmol/kg, i.p.). In the paw skin incision model of post-operative pain, co-administration of NS-9283 similarly induced a 6-fold leftward shift of ABT-594 (ED(50)=26 vs. 153 nmol/kg). In monoiodo-acetate induced knee joint pain, co-administration of NS-9283 enhanced the potency of ABT-594 by 5-fold (ED(50)=1.0 vs. 4.6 nmol/kg). In pharmacological MRI, co-administration of NS-9283 was shown to lead to a leftward shift of ABT-594 dose-response for cortical activation. ABT-594 induced CNS-related adverse effects were not exacerbated in presence of an efficacious dose of NS-9283 (3.5 µmol/kg). Acute challenge of NS-9283 produced no cross sensitization in nicotine-conditioned animals. These results demonstrate that selective positive allosteric modulation at the α4ß2 nAChR potentiates nAChR agonist-induced analgesic activity across neuropathic and nociceptive preclinical pain models without potentiating ABT-594-mediated adverse effects, suggesting that selective positive modulation of α4ß2 nAChR by PAM may represent a novel analgesic approach.


Subject(s)
Analgesics/therapeutic use , Azetidines/therapeutic use , Nicotinic Agonists/therapeutic use , Oxadiazoles/therapeutic use , Pain/drug therapy , Pyridines/therapeutic use , Receptors, Nicotinic/metabolism , Allosteric Regulation , Analgesics/administration & dosage , Analgesics/adverse effects , Animals , Azetidines/administration & dosage , Azetidines/adverse effects , Behavior, Animal/drug effects , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Drug Therapy, Combination , Magnetic Resonance Imaging , Male , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/adverse effects , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Oxadiazoles/administration & dosage , Oxadiazoles/adverse effects , Pain/metabolism , Pyridines/administration & dosage , Pyridines/adverse effects , Rats , Rats, Sprague-Dawley
5.
Biochem Pharmacol ; 81(12): 1374-87, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21219879

ABSTRACT

During the last two decades, functional neuroimaging technology, especially functional magnetic resonance imaging (fMRI), has improved tremendously, with new attention towards resting-state functional connectivity of the brain. This development has allowed scientists to study changes in brain structure and function, and probe these two properties under conditions of evoked stimulation, disease and drug administration. In the domain of functional imaging, the identification and characterization of central nervous system (CNS) functional networks have emerged as potential biomarkers for CNS disorders in humans. Recent attempts to translate clinical neuroimaging methodology to preclinical studies have also been carried out, which offer new opportunities in translational neuroscience research. In this paper, we review recent developments in structural and functional MRI and their use to probe functional connectivity in various CNS disorders such as schizophrenia, mood disorders, Alzheimer's disease (AD) and pain.


Subject(s)
Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/pathology , Central Nervous System/pathology , Magnetic Resonance Imaging , Translational Research, Biomedical/methods , Animals , Brain/pathology , Brain/physiology , Humans
6.
Br J Pharmacol ; 162(2): 428-40, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20880025

ABSTRACT

BACKGROUND AND PURPOSE: Cannabinoid CB2 receptor activation by selective agonists has been shown to produce analgesic effects in preclinical models of inflammatory and neuropathic pain. However, mechanisms underlying CB2-mediated analgesic effects remain largely unknown. The present study was conducted to elucidate the CB2 receptor expression in 'pain relevant' tissues and the potential sites of action of CB2 agonism in rats. EXPERIMENTAL APPROACH: Expression of cannabinoid receptor mRNA was evaluated by quantitative RT-PCR in dorsal root ganglia (DRGs), spinal cords, paws and several brain regions of sham, chronic inflammatory pain (CFA) and neuropathic pain (spinal nerve ligation, SNL) rats. The sites of CB2 mediated antinociception were evaluated in vivo following intra-DRG, intrathecal (i.t.) or intraplantar (i.paw) administration of potent CB2-selective agonists A-836339 and AM1241. KEY RESULTS: CB2 receptor gene expression was significantly up-regulated in DRGs (SNL and CFA), spinal cords (SNL) or paws (CFA) ipsilateral to injury under inflammatory and neuropathic pain conditions. Systemic A-836339 and AM1241 produced dose-dependent efficacy in both inflammatory and neuropathic pain models. Local administration of CB2 agonists also produced significant analgesic effects in SNL (intra-DRG and i.t.) and CFA (intra-DRG) pain models. In contrast to A-836339, i.paw administration of AM-1241 dose-relatedly reversed the CFA-induced thermal hyperalgesia, suggesting that different mechanisms may be contributing to its in vivo properties. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that both DRG and spinal cord are important sites contributing to CB2 receptor-mediated analgesia and that the changes in CB2 receptor expression play a crucial role for the sites of action in regulating pain perception.


Subject(s)
Analgesics/pharmacology , Neuralgia/drug therapy , Pain/drug therapy , Receptor, Cannabinoid, CB2/metabolism , Analgesia , Analgesics/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Male , Neuralgia/chemically induced , Opioid Peptides/metabolism , Pain/metabolism , Pain Perception , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists , Spinal Cord/drug effects , Spinal Cord/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use
7.
Brain Res ; 1369: 158-66, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21047496

ABSTRACT

The TRPV1 receptor functions as a molecular integrator, and blockade of this receptor modulates enhanced somatosensitivity across several animal models of pathological pain, including models of osteoarthritic (OA) pain. In order to further characterize the contributions of TRPV1 to OA-related pain, we investigated the systemic effects of a selective TRPV1 receptor antagonist, A-889425, on grip force behavior, and on the evoked and spontaneous firing of spinal wide dynamic range (WDR) and nociceptive specific (NS) neurons in the monoiodoacetate (MIA) model of OA. Administration of A-889425 (10-300 µmol/kg, p.o.) alleviated grip force impairment in OA rats 3 weeks after the MIA injection. Also at 3 weeks post-MIA injection, the responses of WDR and NS neurons to 300 g von Frey hair stimulation of the knee joint were significantly reduced by A-889425 administration (10 and 30 µmol/kg, i.v.) in OA, but not sham-OA rats. Spontaneous firing of WDR neurons was elevated in the OA rats compared to sham-OA rats and may reflect ongoing discomfort in the OA animal. In addition to an effect on mechanotransmission, systemic administration of A-889425 reduced the elevated spontaneous firing of WDR neurons in OA rats but did not alter spontaneous firing in sham rats. The present data demonstrate that blockade of TRPV1 receptors modulates the firing of two important classes of spinal nociceptive neurons in a rat model of OA. The effect of A-889425 on neuronal responses to intense mechanical stimulation of the knee and on the spontaneous firing of WDR neurons adds to the growing appreciation for the role of TRPV1 receptors in pathological mechanotransmission and possibly non-evoked discomfort, respectively.


Subject(s)
Nociceptors/metabolism , Pain/metabolism , Spinal Cord/metabolism , TRPV Cation Channels/metabolism , Animals , Disease Models, Animal , Electrophysiology , Male , Nociceptors/drug effects , Osteoarthritis/complications , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Pain/physiopathology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , TRPV Cation Channels/drug effects
8.
Brain Res ; 1354: 74-84, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20682302

ABSTRACT

The histamine H(3) receptor is predominantly expressed in the central nervous system and plays a role in diverse physiological mechanisms. In the present study, the effects of GSK189254, a potent and selective H(3) antagonist, were characterized in preclinical pain models in rats. Systemic GSK189254 produced dose-dependent efficacy (ED(50)=0.77 mg/kg i.p.) in a rat model of monoiodoacetate (MIA) induced osteoarthritic (OA) pain as evaluated by hindlimb grip force. The role of H(3) receptors in regulating pain perception was further demonstrated using other structurally distinct H(3) antagonists. GSK189254 also displayed efficacy in a rat surrogate model indicative of central sensitization, namely phase 2 response of formalin-induced flinching, and attenuated tactile allodynia in the spinal nerve ligation model of neuropathic pain (ED(50)=1.5mg/kg i.p.). In addition, GSK189254 reversed persistent (CFA) (ED(50)=2.1mg/kg i.p,), whereas was ineffective in acute (carrageenan) inflammatory pain. When administered intrathecally (i.t.) to the lumbar spinal cord, GSK189254 produced robust effects in relieving the OA pain (ED(50)=0.0027 mg/kg i.t.). The systemic GSK189254 effect was completely reversed by the alpha-adrenergic receptor antagonist phentolamine (i.p. and i.t.) but not by the opioid receptor antagonist naloxone (i.p.). Furthermore, the i.t. GSK189254 effect was abolished when co-administered with phentolamine (i.t.). These results suggest that the spinal cord is an important site of action for H(3) antagonism and the effect can be associated with activation of the noradrenergic system. Our data also provide support that selective H(3) antagonists may represent a class of agents for the treatment of pain disorders.


Subject(s)
Histamine H3 Antagonists/pharmacology , Neurons/drug effects , Norepinephrine/metabolism , Pain Measurement/drug effects , Pain/drug therapy , Receptors, Histamine H3/metabolism , Adrenergic alpha-Antagonists/pharmacology , Analysis of Variance , Animals , Benzazepines/pharmacology , Dose-Response Relationship, Drug , Formaldehyde , Hand Strength , Injections, Spinal , Male , Motor Activity/drug effects , Neurons/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pain/chemically induced , Pain/metabolism , Pain Perception/drug effects , Phentolamine/pharmacology , Rats , Rats, Sprague-Dawley
9.
Pharmacol Biochem Behav ; 95(1): 41-50, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20004681

ABSTRACT

The histamine H(4) receptor (H(4)R) is expressed primarily on cells involved in inflammation and immune responses. To determine the potential role of H(4)R in pain transmission, the effects of JNJ7777120, a potent and selective H(4) antagonist, were characterized in preclinical pain models. Administration of JNJ7777120 fully blocked neutrophil influx observed in a mouse zymosan-induced peritonitis model (ED(50)=17 mg/kg s.c., 95% CI=8.5-26) in a mast cell-dependent manner. JNJ7777120 potently reversed thermal hyperalgesia observed following intraplantar carrageenan injection of acute inflammatory pain (ED(50)=22 mg/kg i.p., 95% CI=10-35) in rats and significantly decreased the myeloperoxide activity in the carrageenan-injected paw. In contrast, no effects were produced by either H(1)R antagonist diphenhydramine, H(2)R antagonists ranitidine, or H(3)R antagonist ABT-239. JNJ7777120 also exhibited robust anti-nociceptive activity in persistent inflammatory (CFA) pain with an ED(50) of 29 mg/kg i.p. (95% CI=19-40) and effectively reversed monoiodoacetate (MIA)-induced osteoarthritic joint pain. This compound also produced dose-dependent anti-allodynic effects in the spinal nerve ligation (ED(50)=60 mg/kg) and sciatic nerve constriction injury (ED(50)=88 mg/kg) models of chronic neuropathic pain, as well as in a skin-incision model of acute post-operative pain (ED(50)=68 mg/kg). In addition, the analgesic effects of JNJ7777120 were maintained following repeated administration and were evident at the doses that did not cause neurologic deficits in rotarod test. Our results demonstrate that selective blockade of H(4) receptors in vivo produces significant anti-nociception in animal models of inflammatory and neuropathic pain.


Subject(s)
Analgesics/pharmacology , Disease Models, Animal , Inflammation/drug therapy , Peripheral Nervous System Diseases/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Analgesics/therapeutic use , Animals , Male , Mice , Mice, Inbred BALB C , Radioligand Assay , Rats , Receptors, Histamine , Receptors, Histamine H4
10.
J Med Chem ; 53(1): 295-315, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19921781

ABSTRACT

Several 3-acylindoles with high affinity for the CB(2) cannabinoid receptor and selectivity over the CB(1) receptor have been prepared. A variety of 3-acyl substituents were investigated, and the tetramethylcyclopropyl group was found to lead to high affinity CB(2) agonists (5, 16). Substitution at the N1-indole position was then examined. A series of aminoalkylindoles was prepared and several substituted aminoethyl derivatives were active (23-27, 5) at the CB(2) receptor. A study of N1 nonaromatic side chain variants provided potent agonists at the CB(2) receptor (16, 35-41, 44-47, 49-54, and 57-58). Several polar side chains (alcohols, oxazolidinone) were well-tolerated for CB(2) receptor activity (41, 50), while others (amide, acid) led to weaker or inactive compounds (55 and 56). N1 aromatic side chains also afforded several high affinity CB(2) receptor agonists (61, 63, 65, and 69) but were generally less potent in an in vitro CB(2) functional assay than were nonaromatic side chain analogues.


Subject(s)
Indoles/pharmacology , Ketones/pharmacology , Receptor, Cannabinoid, CB2/agonists , Drug Design , Humans , Indoles/chemical synthesis , Indoles/chemistry , Ketones/chemical synthesis , Ketones/chemistry , Ligands , Molecular Structure , Receptor, Cannabinoid, CB1/agonists , Stereoisomerism , Structure-Activity Relationship
11.
Behav Brain Res ; 204(1): 77-81, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19464323

ABSTRACT

The pro-inflammatory cytokine interleukin-1beta (IL-1beta) has been implicated in both inflammatory processes and nociceptive neurotransmission. Activation of P2X7 receptors is the mechanism by which ATP stimulates the rapid maturation and release of IL-1beta from macrophages and microglial cells. Recently, selective P2X7 receptor antagonists have been shown to reduce inflammatory and neuropathic pain in animal models. However, the mechanisms underlying these analgesic effects are unknown. The present studies characterize the pharmacology and antinociceptive effects of a structurally novel P2X7 antagonist. A-839977 potently (IC50=20-150 nM) blocked BzATP-evoked calcium influx at recombinant human, rat and mouse P2X7 receptors. A-839977 also potently blocked agonist-evoked YO-PRO uptake and IL-1beta release from differentiated human THP-1 cells. Systemic administration of A-839977 dose-dependently reduced thermal hyperalgesia produced by intraplantar administration of complete Freund's adjuvant (CFA) (ED50=100 micromol/kg, i.p.) in rats. A-839977 also produced robust antihyperalgesia in the CFA model of inflammatory pain in wild-type mice (ED50=40 micromol/kg, i.p.), but the antihyperalgesic effects of A-839977 were completely absent in IL-1alphabeta knockout mice. These data demonstrate that selective blockade of P2X7 receptors in vivo produces significant antinociception in animal models of inflammatory pain and suggest that the antihyperalgesic effects of P2X7 receptor blockade in an inflammatory pain model in mice are mediated by blocking the release of IL-1beta.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Pain/drug therapy , Pyridines/pharmacology , Receptors, Purinergic P2/metabolism , Tetrazoles/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Analgesics, Non-Narcotic/chemistry , Animals , Calcium/metabolism , Cell Line , Dose-Response Relationship, Drug , Freund's Adjuvant , Hot Temperature , Humans , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Pain/chemically induced , Pain/metabolism , Purinergic P2 Receptor Antagonists , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2X7 , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Tetrazoles/chemistry
12.
Eur J Pharmacol ; 613(1-3): 39-45, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19376109

ABSTRACT

This study was conducted to characterize movement-induced pain in a rat model of knee joint osteoarthritis and validate this behavioral assessment by evaluating the effects of clinically used analgesic compounds. Unilateral intra-articular administration of a chondrocyte glycolytic inhibitor monoiodoacetate, was used to induce knee joint osteoarthritis in Sprague-Dawley rats. In this osteoarthritis model, histologically erosive disintegration of the articular surfaces of the ipsilateral joint are observed which closely mimic the clinical picture of osteoarthritis. Movement-induced pain behavior was measured using hind limb compressive grip force evaluation. The animals exhibited pain behaviors epitomized by a long-lasting decrement in bilateral compressive hind limb grip force following unilateral knee injury. The effects of clinically used reference analgesics were evaluated 20 days following i.a. injection of monoiodoacetate. Full analgesic activity was observed for tramadol, celecoxib and diclofenac; moderate effects for indomethacin, duloxetine and gabapentin but weak or no effects for acetaminophen, ibuprofen and lamotrigine. As morphine reduced grip force in naïve rats, its analgesic effects could not be accurately evaluated in this model. Finally, the effects of celecoxib were maintained following chronic dosing. The results indicate that this in vivo model utilizing a movement-induced pain behavior spawned by knee joint osteoarthritis may provide a valuable tool in examining the role of potential analgesic targets in osteoarthritic pain. As the model is clinically relevant, it will further enhance the mechanistic understanding of chronic arthritic joint pain and help in developing newer and better therapeutic strategies to manage osteoarthritis pain.


Subject(s)
Analgesics/pharmacology , Movement , Osteoarthritis/drug therapy , Pain/drug therapy , Pain/etiology , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Analgesics/therapeutic use , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Male , Osteoarthritis/complications , Pain/complications , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Time Factors
13.
Pain ; 142(1-2): 27-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135797

ABSTRACT

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel that functions as an integrator of multiple pain stimuli including heat, acid, capsaicin and a variety of putative endogenous lipid ligands. TRPV1 antagonists have been shown to decrease inflammatory pain in animal models and to produce limited hyperthermia at analgesic doses. Here, we report that ABT-102, which is a potent and selective TRPV1 antagonist, is effective in blocking nociception in rodent models of inflammatory, post-operative, osteoarthritic, and bone cancer pain. ABT-102 decreased both spontaneous pain behaviors and those evoked by thermal and mechanical stimuli in these models. Moreover, we have found that repeated administration of ABT-102 for 5-12 days increased its analgesic activity in models of post-operative, osteoarthritic, and bone cancer pain without an associated accumulation of ABT-102 concentration in plasma or brain. Similar effects were also observed with a structurally distinct TRPV1 antagonist, A-993610. Although a single dose of ABT-102 produced a self-limiting increase in core body temperature that remained in the normal range, the hyperthermic effects of ABT-102 effectively tolerated following twice-daily dosing for 2 days. Therefore, the present data demonstrate that, following repeated administration, the analgesic activity of TRPV1 receptor antagonists is enhanced, while the associated hyperthermic effects are attenuated. The analgesic efficacy of ABT-102 supports its advancement into clinical studies.


Subject(s)
Analgesics/administration & dosage , Fever/drug therapy , Indazoles/administration & dosage , Pain Threshold/drug effects , Pain/drug therapy , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Animals , Body Temperature/drug effects , Bone Neoplasms/complications , Calcium/metabolism , Disease Models, Animal , Drug Interactions , Fever/chemically induced , Inflammation/complications , Male , Mice , Mice, Inbred C3H , Motor Activity/drug effects , Osteoarthritis/complications , Pain/etiology , Pain Measurement , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors , Urea/administration & dosage
14.
J Pharmacol Exp Ther ; 328(1): 141-51, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18931146

ABSTRACT

Studies demonstrating the antihyperalgesic and antiallodynic effects of cannabinoid CB(2) receptor activation have been largely derived from the use of receptor-selective ligands. Here, we report the identification of A-836339 [2,2,3,3-tetramethyl-cyclopropanecarboxylic acid [3-(2-methoxy-ethyl)-4,5-dimethyl-3H-thiazol-(2Z)-ylidene]-amide], a potent and selective CB(2) agonist as characterized in in vitro pharmacological assays and in in vivo models of pain and central nervous system (CNS) behavior models. In radioligand binding assays, A-836339 displays high affinities at CB(2) receptors and selectivity over CB(1) receptors in both human and rat. Likewise, A-836339 exhibits high potencies at CB(2) and selectivity over CB(1) receptors in recombinant fluorescence imaging plate reader and cyclase functional assays. In addition A-836339 exhibits a profile devoid of significant affinity at other G-protein-coupled receptors and ion channels. A-836339 was characterized extensively in various animal pain models. In the complete Freund's adjuvant model of inflammatory pain, A-836339 exhibits a potent CB(2) receptor-mediated antihyperalgesic effect that is independent of CB(1) or mu-opioid receptors. A-836339 has also demonstrated efficacies in the chronic constrain injury (CCI) model of neuropathic pain, skin incision, and capsaicin-induced secondary mechanical hyperalgesia models. Furthermore, no tolerance was developed in the CCI model after subchronic treatment with A-836339 for 5 days. In assessing CNS effects, A-836339 exhibited a CB(1) receptor-mediated decrease of spontaneous locomotor activities at a higher dose, a finding consistent with the CNS activation pattern observed by pharmacological magnetic resonance imaging. These data demonstrate that A-836339 is a useful tool for use of studying CB(2) receptor pharmacology and for investigation of the role of CB(2) receptor modulation for treatment of pain in preclinical animal models.


Subject(s)
Amides/pharmacology , Cyclopropanes/pharmacology , Inflammation/physiopathology , Pain/physiopathology , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Dermatologic Surgical Procedures , Hindlimb , Humans , Hyperalgesia/physiopathology , Kidney/embryology , Magnetic Resonance Imaging/methods , Male , Pain, Postoperative/physiopathology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists
15.
J Med Chem ; 51(22): 7094-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18983139

ABSTRACT

cis-4-(Piperazin-1-yl)-5,6,7a,8,9,10,11,11a-octahydrobenzofuro[2,3-h]quinazolin-2-amine, 4 (A-987306) is a new histamine H(4) antagonist. The compound is potent in H(4) receptor binding assays (rat H(4), K(i) = 3.4 nM, human H(4) K(i) = 5.8 nM) and demonstrated potent functional antagonism in vitro at human, rat, and mouse H(4) receptors in cell-based FLIPR assays. Compound 4 also demonstrated H(4) antagonism in vivo in mice, blocking H(4)-agonist induced scratch responses, and showed anti-inflammatory activity in mice in a peritonitis model. Most interesting was the high potency and efficacy of this compound in blocking pain responses, where it showed an ED(50) of 42 mumol/kg (ip) in a rat post-carrageenan thermal hyperalgesia model of inflammatory pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzofurans/pharmacology , Hyperalgesia/drug therapy , Pain/prevention & control , Quinazolines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Carrageenan , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Humans , Hyperalgesia/chemically induced , Ligands , Mice , Molecular Structure , Pain/physiopathology , Peritonitis/drug therapy , Quinazolines/chemical synthesis , Quinazolines/chemistry , Rats , Receptors, Histamine , Receptors, Histamine H4 , Stereoisomerism , Structure-Activity Relationship
16.
J Med Chem ; 51(6): 1904-12, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18311894

ABSTRACT

A series of potent indol-3-yl-tetramethylcyclopropyl ketones have been prepared as CB 2 cannabinoid receptor ligands. Two unsubstituted indoles ( 5, 32) were the starting points for an investigation of the effect of indole ring substitutions on CB 2 and CB 1 binding affinities and activity in a CB 2 in vitro functional assay. Indole ring substitutions had varying effects on CB 2 and CB 1 binding, but were generally detrimental to agonist activity. Substitution on the indole ring did lead to improved CB 2/CB 1 binding selectivity in some cases (i.e., 7- 9, 15- 20). All indoles with the morpholino-ethyl side chain ( 32- 43) exhibited weaker binding affinity and less agonist activity relative to that of their tetrahydropyranyl-methyl analogs ( 5- 31). Several agonists were active in the complete Freund's adjuvant model of chronic inflammatory thermal hyperalgesia ( 32, 15).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Indoles/pharmacology , Ketones/pharmacology , Receptor, Cannabinoid, CB2/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Binding, Competitive , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Indoles/chemical synthesis , Indoles/chemistry , Ketones/chemical synthesis , Ketones/chemistry , Ligands , Molecular Conformation , Rats , Receptor, Cannabinoid, CB1/drug effects , Stereoisomerism , Structure-Activity Relationship
17.
J Pharmacol Exp Ther ; 319(3): 1376-85, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16982702

ABSTRACT

ATP-sensitive P2X(7) receptors are localized on cells of immunological origin including glial cells in the central nervous system. Activation of P2X(7) receptors leads to rapid changes in intracellular calcium concentrations, release of the proinflammatory cytokine interleukin-1beta (IL-1beta), and following prolonged agonist exposure, cytolytic plasma membrane pore formation. P2X(7) knockout mice show reduced inflammation as well as decreased nociceptive sensitivity following peripheral nerve injury. A-740003 (N-(1-{[(cyanoimino)(5-quinolinylamino) methyl] amino}-2,2-dimethylpropyl)-2-(3,4-dimethoxyphenyl)acetamide) is a novel competitive antagonist of P2X(7) receptors (IC(50) values = 40 nM for human and 18 nM for rat) as measured by agonist-stimulated changes in intracellular calcium concentrations. A-740003 showed weak or no activity (IC(50) > 10 muM) at other P2 receptors and an array of other neurotransmitter and peptide receptors, ion channels, reuptake sites, and enzymes. A-740003 potently blocked agonist-evoked IL-1beta release (IC(50) = 156 nM) and pore formation (IC(50) = 92 nM) in differentiated human THP-1 cells. Systemic administration of A-740003 produced dose-dependent antinociception in a spinal nerve ligation model (ED(50) = 19 mg/kg i.p.) in the rat. A-740003 also attenuated tactile allodynia in two other models of neuropathic pain, chronic constriction injury of the sciatic nerve and vincristine-induced neuropathy. In addition, A-740003 effectively reduced thermal hyperalgesia observed following intraplantar administration of carrageenan or complete Freund's adjuvant (ED(50) = 38-54 mg/kg i.p.). A-740003 was ineffective in attenuating acute thermal nociception in normal rats and did not alter motor performance at analgesic doses. These data demonstrate that selective blockade of P2X(7) receptors in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


Subject(s)
Acetamides/pharmacology , Analgesics , Pain/drug therapy , Pain/etiology , Peripheral Nervous System Diseases/complications , Purinergic P2 Receptor Antagonists , Quinolines/pharmacology , Animals , Antineoplastic Agents, Phytogenic/toxicity , Calcium/metabolism , Cell Line , Coloring Agents , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Freund's Adjuvant/pharmacology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/complications , Inflammation/drug therapy , Interleukin-1beta/metabolism , Male , Motor Activity/drug effects , Nociceptors/drug effects , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X7 , Sciatic Neuropathy/prevention & control , Spinal Nerves/injuries , Vincristine/toxicity
18.
Pain ; 102(1-2): 195-201, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12620611

ABSTRACT

The analgesia produced by low and high frequency transcutaneous electrical nerve stimulation (TENS) is mediated by the release of mu- or delta-opioids, respectively in the central nervous system. Repeated administration of either mu- or delta-opioid agonists induce opioid analgesic tolerance. Thus, we tested if repeated administration of TENS (either low or high frequency) in rats leads to a development of tolerance to its antihyperalgesic effects with a corresponding cross-tolerance to mu- and delta-opioid agonists. Unilateral knee joint inflammation (3% carrageenan) was induced in adult Sprague-Dawley rats. Either low (4 Hz) or high frequency (100 Hz) TENS was administered for 6 days (20 min daily) to the inflamed knee joint under halothane anesthesia. The no TENS controls were administered anesthesia only for the same period. Withdrawal threshold to mechanical stimuli was measured before and after administration of TENS on each day and also on the sixth day. A separate group of animals was tested for tolerance to either the mu-opioid agonist, morphine (1.32, 3.95, 13.2 nmol/10 ml, intrathecal (i.t.)) or the delta-opioid agonist, SNC-80 (6, 20, 60, 120 nmol/10 ml, i.t.) 30 min after i.t. administration. The reduced mechanical withdrawal threshold following the induction of inflammation was reversed by the application of TENS. However, repeatedly administering either low or high frequency TENS for 6 days, lead to a diminution in its effectiveness in reversing the ipsilateral secondary mechanical hyperalgesia by the fourth day. The effects of morphine in the low and SNC-80 in the high frequency TENS groups were significantly less than the group that did not receive TENS. On the other hand, morphine and SNC-80 were similar to the no TENS control in the high and low frequency TENS groups, respectively. Thus, repeated administration of low and high frequency TENS leads to a development of opioid tolerance with a corresponding cross-tolerance to i.t. administered mu- and delta-opioid agonists, respectively. Clinically, it can be inferred that a treatment schedule of repeated daily TENS administration should be avoided to possibly obviate the induction of tolerance.


Subject(s)
Drug Tolerance/physiology , Morphine/therapeutic use , Pain/drug therapy , Transcutaneous Electric Nerve Stimulation/methods , Animals , Benzamides/administration & dosage , Benzamides/therapeutic use , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/drug therapy , Knee Joint/drug effects , Knee Joint/physiopathology , Male , Morphine/administration & dosage , Pain Measurement , Physical Stimulation , Piperazines/administration & dosage , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Transcutaneous Electric Nerve Stimulation/adverse effects
19.
Pain ; 100(1-2): 183-90, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12435471

ABSTRACT

Transcutaneous electrical nerve stimulation (TENS) partially reduces primary hyperalgesia and is frequency dependent such that high frequency TENS produces approximately a 30% reduction in hyperalgesia whereas low frequency TENS has no effect. Both high and low frequency TENS completely reduce secondary hyperalgesia by activation of mu and delta- opioid receptors in the spinal cord and rostral-ventral medulla suggesting an opiate mediated analgesia. Clonidine in combination with opiates produces a synergistic interaction such that there is a potentiated reduction in hyperalgesia. Thus, we tested if combined application of clonidine with TENS would enhance the reduction in primary hyperalgesia. Male Sprague-Dawley rats were inflamed by subcutaneous injection of 3% carrageenan into one hindpaw. Withdrawal latency to radiant heat and withdrawal threshold to mechanical stimuli were assessed before and after inflammation and after administration of clonidine (0.002-2 mg/kg, intraperitoneal (i.p.)) with either low (4 Hz) or high (100 Hz) frequency TENS. Clonidine alone reduced both heat and mechanical hyperalgesia with ED50s of 0.02 and 1.0 mg/kg, respectively. In combination with either low or high frequency TENS, the dose-response curve shifted to the left and was significantly different from clonidine alone. The ED50s for heat and mechanical hyperalgesia following low frequency TENS with clonidine were 0.002 and 0.2 mg/kg, respectively and those following high frequency TENS with clonidine were 0.005 and 0.15 mg/kg, respectively. Thus, combined use of clonidine and TENS enhances the reduction in analgesia produced by TENS and enhances the potency of clonidine. It would thus be expected that one would reduce the side effects of clonidine and enhance analgesic efficacy with combinations of pharmaceutical and non-pharmaceutical treatments.


Subject(s)
Analgesics/pharmacology , Clonidine/pharmacology , Hyperalgesia/drug therapy , Transcutaneous Electric Nerve Stimulation , Animals , Carrageenan , Combined Modality Therapy , Inflammation/chemically induced , Male , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...