pregabalin has been researched along with Disease-Models--Animal* in 133 studies
6 review(s) available for pregabalin and Disease-Models--Animal
Article | Year |
---|---|
Patients with POAG have lower corneal endothelial cell density than healthy controls of the same age. This may be attributed to mechanical damage from elevated IOP and toxicity of glaucoma medications.. Mycophenolic acid was detected in all cats. The dose 10 mg/kg given q12h for 1 week was tolerated (n = 3). The efficacy of MMF as an immunosuppressant and long-term safety in cats of this dosage regimen is unknown.. T Topics: Acetylcholine; Acinetobacter baumannii; Actinobacteria; Action Potentials; Adalimumab; Adaptation, Physiological; Adipates; Administration, Oral; Adolescent; Adrenal Glands; Adsorption; Adult; Aged; Aged, 80 and over; Aging; AIDS-Related Opportunistic Infections; Aldosterone; Amino Acids; Ammonia; Amoxicillin; AMP-Activated Protein Kinases; Animals; Antacids; Anti-Bacterial Agents; Antineoplastic Agents; Antirheumatic Agents; Apgar Score; Area Under Curve; ARNTL Transcription Factors; Arterial Pressure; Arthritis, Juvenile; Athletes; Attention; Biodegradation, Environmental; Biofilms; Biofuels; Biological Therapy; Biomass; Biomimetic Materials; Bioreactors; Birth Weight; Bismuth; Blood Flow Velocity; Bone and Bones; Brain Injuries, Traumatic; Calcium; Calcium Channels; Capsaicin; Carbon; Carcinoma, Hepatocellular; Cardiomegaly, Exercise-Induced; Cartilage; Cartilage, Articular; Case-Control Studies; Catalysis; Cats; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cell Death; Cell Differentiation; Cell Line, Tumor; Cell Membrane; Charcoal; Chemokine CCL2; Child; Child, Preschool; Chondrogenesis; Chronic Disease; Circadian Clocks; Circadian Rhythm Signaling Peptides and Proteins; Clarithromycin; Coccidioides; Coccidioidomycosis; Cognitive Behavioral Therapy; Coinfection; Color; Coloring Agents; Computer Simulation; Computers, Molecular; Consensus; Corticosterone; Cyclic AMP Response Element-Binding Protein; Cytochrome P-450 Enzyme System; Death, Sudden, Cardiac; Density Functional Theory; Diabetes Mellitus, Type 2; Diabetic Retinopathy; Dialysis Solutions; Disease Models, Animal; Dogs; Dopamine Agonists; Dose-Response Relationship, Drug; Doxorubicin; Drug Administration Schedule; Drug Resistance, Bacterial; Drug Therapy, Combination; Electrocardiography; Electrocardiography, Ambulatory; Electrolytes; Endocardium; Endocrine Disruptors; Endocytosis; Endoscopy, Gastrointestinal; Escherichia coli Proteins; Esters; Evolution, Molecular; Executive Function; Feasibility Studies; Female; Ferric Compounds; Fluorescence; Fluorescent Dyes; Fluorine Radioisotopes; Frailty; Free Radical Scavengers; Gabapentin; Geriatric Assessment; Glucaric Acid; Glucocorticoids; Glucose; Glucose Metabolism Disorders; Halogenated Diphenyl Ethers; Heart Rate; Heart Ventricles; HEK293 Cells; Helicobacter Infections; Helicobacter pylori; Hep G2 Cells; Hepatocytes; Humans; Hungary; Hydrogen Sulfide; Hydrogen-Ion Concentration; Immunologic Factors; Immunomodulation; Immunosuppressive Agents; Independent Living; Indocyanine Green; Infant; Infant Formula; Infant Mortality; Infant, Newborn; Infant, Newborn, Diseases; Inflorescence; Insulin Resistance; Insulins; International Agencies; Iron; Isotonic Solutions; Kidney Failure, Chronic; Kinetics; Lactones; Leukocytes, Mononuclear; Liver Neoplasms; Macular Edema; Magnetic Resonance Imaging; Magnetic Resonance Spectroscopy; Magnetosomes; Male; Medical Audit; Mesenchymal Stem Cells; Metabolic Syndrome; Metformin; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Nude; Middle Aged; Molecular Conformation; Molecular Targeted Therapy; Motor Activity; Multiple Sclerosis; Mycophenolic Acid; Netherlands; Neuropsychological Tests; Nuclear Energy; Organs at Risk; Osteoarthritis; Osteoarthritis, Hip; Oxidation-Reduction; Palladium; Pericardium; Perinatal Death; Peritoneal Dialysis; Phantoms, Imaging; Pharmaceutical Preparations; Phospholipids; Phosphorylation; Physical Conditioning, Human; Physical Endurance; Pilot Projects; Polyketides; Polymers; Positron-Emission Tomography; Postoperative Period; Potassium; Powders; Pramipexole; Predictive Value of Tests; Pregabalin; Pregnancy; Pregnancy Outcome; Protein Structure, Secondary; Proton Pump Inhibitors; Puberty; Pulmonary Circulation; Quality Assurance, Health Care; Quantum Dots; Radiometry; Radiotherapy Dosage; Radiotherapy Planning, Computer-Assisted; Radiotherapy, Intensity-Modulated; Rats, Sprague-Dawley; Receptors, CCR2; Receptors, Transferrin; Regeneration; Registries; Renal Insufficiency, Chronic; Reproducibility of Results; Research Design; Restless Legs Syndrome; Retina; Retinoid X Receptor alpha; Retrospective Studies; Rhenium; Risk Factors; RNA, Messenger; Severity of Illness Index; Sex Factors; Sodium; Sodium Fluoride; Solvents; Spectrometry, Fluorescence; Spectroscopy, Fourier Transform Infrared; Stereoisomerism; Stroke; Structure-Activity Relationship; Tachycardia, Ventricular; Tetracycline; Tetrahydrofolate Dehydrogenase; Tetrahydronaphthalenes; Thermodynamics; Thiophenes; Time Factors; Tinidazole; Tomography, Optical Coherence; Tomography, X-Ray Computed; Topiramate; Toxoplasma; Toxoplasmosis, Cerebral; Transferrin; Treatment Outcome; Up-Regulation; Upper Extremity; Uremia; Uveitis; Vascular Remodeling; Ventricular Fibrillation; Ventricular Function, Left; Ventricular Function, Right; Ventricular Remodeling; Verapamil; Veterans; Visual Acuity; Vitrectomy; Water Pollutants, Chemical; Zea mays; Zirconium | 2019 |
Analgesic mechanisms of gabapentinoids and effects in experimental pain models: a narrative review.
The focus of perioperative pain management should be to attempt to minimise the nociceptive input and reduce the risk of transition to central sensitisation. Gabapentinoids are being increasingly used as adjuncts for management of perioperative pain. Although gabapentinoids are classed as calcium channel blockers, their mechanisms of action are poorly understood. The analgesic effect in neuropathic pain is well evidenced but the role in postoperative pain is less certain. Medline and EMBASE database searches were conducted to identify studies relating to mechanisms of action and effects in experimental animal models of inflammatory and postoperative pain and human models of experimental pain. The effects of gabapentinoids may be attributed to depression of dorsal horn sensitivity through a multitude of mechanisms. They inhibit calcium mediated neurotransmitter release through effects on α2δ-1 subunits. They inhibit forward trafficking of α2δ-1 from the dorsal root ganglion, their recycling from endosomal compartments, thrombospondin mediated processes and stimulate glutamate uptake by excitatory amino acid transporters. Mechanisms not directly related to neurotransmitter release at dorsal horn include inhibition of descending serotonergic facilitation, stimulation of descending inhibition, anti-inflammatory actions, and influence on the affective component of pain. Gabapentinoids are effective analgesics in most animal models of inflammation and postoperative pain but effects in human models are variable. Topics: Analgesics; Animals; Calcium Channels; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Humans; Neuralgia; Pain, Postoperative; Pregabalin | 2018 |
[Pregabalin and postoperative hyperalgesia. A review].
Numerous studies support the theory that pregabalin causes an antihyperalgesic effect, which could be potentially beneficial in a perioperative setting. By binding to calcium channels pregabalin reduces the release of excitatory neurotransmitters and therefore inhibits central sensitization. Animal studies clearly demonstrated the antihyperalgesic potency of pregabalin but human experiments are, however, inconclusive. Clinical studies with quantitative sensory testing have not yet been published. Although strongly supported by theoretical considerations the routine preoperative application of pregabalin for the prevention of hyperalgesia cannot be recommended due to the lack of clinical studies. Future studies should incorporate secondary hyperalgesia and allodynia as primary parameters. Topics: Analgesics; Animals; Calcium Channels; Disease Models, Animal; gamma-Aminobutyric Acid; Humans; Hyperalgesia; Pain, Postoperative; Pregabalin; Premedication | 2011 |
Pharmacology and mechanism of action of pregabalin: the calcium channel alpha2-delta (alpha2-delta) subunit as a target for antiepileptic drug discovery.
Pregabalin (Lyrica) is a new antiepileptic drug that is active in animal seizure models. Pregabalin is approved in US and Europe for adjunctive therapy of partial seizures in adults, and also has been approved for the treatment of pain from diabetic neuropathy or post-herpetic neuralgia in adults. Recently, it has been approved for treatment of anxiety disorders in Europe. Pregabalin is structurally related to the antiepileptic drug gabapentin and the site of action of both drugs is similar, the alpha2-delta (alpha2-delta) protein, an auxiliary subunit of voltage-gated calcium channels. Pregabalin subtly reduces the synaptic release of several neurotransmitters, apparently by binding to alpha2-delta subunits, and possibly accounting for its actions in vivo to reduce neuronal excitability and seizures. Several studies indicate that the pharmacology of pregabalin requires binding to alpha2-delta subunits, including structure-activity analyses of compounds binding to alpha2-delta subunits and pharmacology in mice deficient in binding at the alpha2-delta Type 1 protein. The preclinical findings to date are consistent with a mechanism that may entail reduction of abnormal neuronal excitability through reduced neurotransmitter release. This review addresses the preclinical pharmacology of pregabalin, and also the biology of the high affinity binding site, and presumed site of action. Topics: Amines; Amino Acid Sequence; Animals; Anticonvulsants; Calcium Channels; Cyclohexanecarboxylic Acids; Disease Models, Animal; Epilepsies, Partial; Epilepsy, Tonic-Clonic; Gabapentin; gamma-Aminobutyric Acid; Molecular Sequence Data; Pregabalin; Protein Conformation | 2007 |
[A new aspect in the research on antiepileptic drugs].
Topics: Acetamides; Amines; Animals; Anticonvulsants; Benzodiazepines; Carbamates; Cyclohexanecarboxylic Acids; Disease Models, Animal; Drug Design; Gabapentin; gamma-Aminobutyric Acid; Humans; Lacosamide; Levetiracetam; Phenylenediamines; Piracetam; Pregabalin; Pregnanolone; Pyrrolidinones; Triazoles | 2007 |
Pregabalin pharmacology and its relevance to clinical practice.
Pregabalin is a potent ligand for the alpha-2-delta subunit of voltage-gated calcium channels in the central nervous system that exhibits potent anticonvulsant, analgesic, and anxiolytic activity in a range of animal models. In addition, pregabalin has been shown to be a highly effective adjunctive therapy for partial seizures in clinical trials. Potent binding to the alpha-2-delta site reduces depolarization-induced calcium influx with a consequential modulation in excitatory neurotransmitter release. Pregabalin has no demonstrated effects on GABAergic mechanisms. Pregabalin demonstrates highly predictable and linear pharmacokinetics, a profile that makes it easy to use in clinical practice. Absorption is extensive, rapid, and proportional to dose. Time to maximal plasma concentration is approximately 1 h and steady state is achieved within 24-48 h. These characteristics reflect the observed onset of efficacy as early as day two in clinical trials. High bioavailability, a mean elimination half life (t(1/2)) of 6.3 h, and dose-proportional maximal plasma concentrations and total exposures predict a dose-response relationship in clinical practice and allow an effective starting dose of 150 mg/day in clinical practice without need for titration. Administration with food has no clinically relevant effect on the amount of pregabalin absorbed, providing for a dosing regimen uncomplicated by meals. Pregabalin does not bind to plasma proteins and is excreted virtually unchanged (<2% metabolism) by the kidneys. It is not subject to hepatic metabolism and does not induce or inhibit liver enzymes such as the cytochrome P450 system. Therefore, pregabalin is unlikely to cause, or be subject to, pharmacokinetic drug-drug interactions--an expectation that has been confirmed in clinical pharmacokinetic studies. However, dose adjustment may be necessary in patients with renal insufficiency. Thus, the pharmacological and pharmacokinetic profiles of pregabalin provide a predictable basis for its use in clinical practice. Topics: Animals; Anticonvulsants; Biological Availability; Calcium Channels; Clinical Trials as Topic; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Schedule; Drug Interactions; Epilepsy; gamma-Aminobutyric Acid; Half-Life; Humans; Pregabalin; Treatment Outcome | 2004 |
127 other study(ies) available for pregabalin and Disease-Models--Animal
Article | Year |
---|---|
Heme oxygenase-1 in the spinal cord plays crucial roles in the analgesic effects of pregabalin and gabapentin in a spared nerve-injury mouse model.
Neuropathic pain remains one of the most intractable types of pain; although calcium channel α. Neuropathic pain was induced by spared nerve injury (SNI) of the sciatic nerve. The mechanical threshold was tested using the von Frey filaments. The expression of spinal glial markers or other genes was examined using reverse transcription polymerase chain reaction.. Systemic HO-1 inhibition reversed the mechanical antiallodynic effects of pregabalin and gabapentin, although peripheral HO-1 inhibition did not alter the mechanical antiallodynic effects of either pregabalin or gabapentin. Intrathecal injection of SnPP or ZnPP abolished the mechanical antiallodynic effects of pregabalin and gabapentin. Pregabalin and gabapentin increased HO-1, arginase-1, and endogenous opioid precursor preproenkephalin gene expression and decreased the expression of glial markers, interleukin-1β, and inducible nitric oxide synthase.. This study suggests that spinal HO-1 plays a crucial role in the analgesic effects of calcium channel α Topics: Analgesics; Animals; Disease Models, Animal; Gabapentin; Heme Oxygenase-1; Male; Mice; Mice, Inbred C57BL; Neuralgia; Pregabalin; Sciatic Nerve; Spinal Cord | 2022 |
Pathogenic Mechanism of Dry Eye-Induced Chronic Ocular Pain and a Mechanism-Based Therapeutic Approach.
Dry eye-induced chronic ocular pain is also called ocular neuropathic pain. However, details of the pathogenic mechanism remain unknown. The purpose of this study was to elucidate the pathogenic mechanism of dry eye-induced chronic pain in the anterior eye area and develop a pathophysiology-based therapeutic strategy.. We used a rat dry eye model with lacrimal gland excision (LGE) to elucidate the pathogenic mechanism of ocular neuropathic pain. Corneal epithelial damage, hypersensitivity, and hyperalgesia were evaluated on the LGE side and compared with the sham surgery side. We analyzed neuronal activity, microglial and astrocytic activity, α2δ-1 subunit expression, and inhibitory interneurons in the trigeminal nucleus. We also evaluated the therapeutic effects of ophthalmic treatment and chronic pregabalin administration on dry eye-induced ocular neuropathic pain.. Dry eye caused hypersensitivity and hyperalgesia on the LGE side. In the trigeminal nucleus of the LGE side, neuronal hyperactivation, transient activation of microglia, persistent activation of astrocytes, α2δ-1 subunit upregulation, and reduced numbers of inhibitory interneurons were observed. Ophthalmic treatment alone did not improve hyperalgesia. In contrast, continuous treatment with pregabalin effectively ameliorated hypersensitivity and hyperalgesia and normalized neural activity, α2δ-1 subunit upregulation, and astrocyte activation.. These results suggest that dry eye-induced hypersensitivity and hyperalgesia are caused by central sensitization in the trigeminal nucleus with upregulation of the α2δ-1 subunit. Here, we showed that pregabalin is effective for treating dry eye-induced ocular neuropathic pain even after chronic pain has been established. Topics: Administration, Ophthalmic; Analgesics; Animals; Astrocytes; Calcium Channels, L-Type; Chronic Disease; Cornea; Disease Models, Animal; Dry Eye Syndromes; Eye Pain; Hyaluronic Acid; Hyperalgesia; Male; Microglia; Neuralgia; Neurons; Ophthalmic Solutions; Pregabalin; Rats; Rats, Sprague-Dawley; Trigeminal Nerve | 2022 |
Effect of caffeine on the anticonvulsant action of pregabalin against electroconvulsions in mice.
Experimental data indicate that caffeine (CAF) can reduce the anticonvulsant activity of antiepileptic drugs (AEDs) in animal models of seizures. The purpose of the current study was to examine the effect of CAF on the protective action of pregabalin (PGB) against electroconvulsions in mice.. Maximal electroshock seizure (MES) test was used in the current study. In addition, the combined treatment with CAF and PGB was assessed in the passive avoidance task (long-term memory) and the chimney test (motor coordination). Drugs were injected intraperitoneally (ip) as single injections. CAF was administered at doses reported to compromise the anticonvulsant action of AEDs in mice.. CAF at a dose of 23.1 mg/kg reduced the anticonvulsant action of PGB in the MES test. The brain concentration of PGB was not significantly changed by CAF and vice versa. In the chimney test, CAF (23.1 mg/kg) protected mice against PGB-induced motor coordination impairment.. Regarding seizure control, it might be suggested that patients with epilepsy treated with PGB should avoid taking CAF. The estimated total brain concentration of PGB and CAF does not suggest a pharmacokinetic interaction as an explanation for these results. Topics: Animals; Anticonvulsants; Brain; Caffeine; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Electroshock; Humans; Mice; Pregabalin; Seizures | 2022 |
Long-lasting reflexive and nonreflexive pain responses in two mouse models of fibromyalgia-like condition.
Nociplastic pain arises from altered nociception despite no clear evidence of tissue or somatosensory system damage, and fibromyalgia syndrome can be highlighted as a prototype of this chronic pain subtype. Currently, there is a lack of effective treatments to alleviate both reflexive and nonreflexive pain responses associated with fibromyalgia condition, and suitable preclinical models are needed to assess new pharmacological strategies. In this context, although in recent years some remarkable animal models have been developed to mimic the main characteristics of human fibromyalgia, most of them show pain responses in the short term. Considering the chronicity of this condition, the present work aimed to develop two mouse models showing long-lasting reflexive and nonreflexive pain responses after several reserpine (RIM) or intramuscular acid saline solution (ASI) injections. To our knowledge, this is the first study showing that RIM6 and ASI mouse models show reflexive and nonreflexive responses up to 5-6 weeks, accompanied by either astro- or microgliosis in the spinal cord as pivotal physiopathology processes related to such condition development. In addition, acute treatment with pregabalin resulted in reflexive pain response alleviation in both the RIM6 and ASI models. Consequently, both may be considered suitable experimental models of fibromyalgia-like condition, especially RIM6. Topics: Animals; Chronic Pain; Disease Models, Animal; Fibromyalgia; Mice; Pregabalin; Reserpine | 2022 |
Co-Combination of Pregabalin and
The current study reports the fabrication of co-combination gel using Pregabalin and Withania coagulans fruit extract to validate its effectiveness for neuropathic pain in chronic constriction injury (CCI) rat models. Three topical gels were prepared using Carbopol 934 through a pseudo-ternary phase diagram incorporating the Pregabalin (2.5%), Withania coagulans extract (2%), and co-combination of both Pregabalin (2.5%) and Withania coagulans extract (2%). Gels were characterized. FTIR showed a successful polymeric network of the gel without any interaction. The drug distribution at the molecular level was confirmed by XRD. The AFM images topographically indicated the rough surface of gels with a size range from 0.25 to 330 nm. DSC showed the disappearance of sharp peaks of the drug and extract, showing successful incorporation into the polymeric network of gels. The in vitro drug release of co-combination gel was 73% over 48 h. The mechanism of drug release by combination gel was Higuchi+ fickian with values of n (0.282) and R2 (0.947). An in vivo study for pain assessment via four methods: (i) heat hyperalgesia, (ii) cold allodynia, (iii) mechano-hyperalgesia, and (iv) dynamic mechano-allodynia, confirmed that topical treatment with co-combination gel reduced the pain significantly as indicated by the p value: R1 (p < 0.001), R2 (p < 0.001), R3 (p < 0.015), and R4 (p < 0.0344). The significance order was R2 (****) > R1 (***) > R3 (**) > R4 (*) > R5 (ns). Topics: Animals; Constriction; Disease Models, Animal; Gels; Hyperalgesia; Neuralgia; Peripheral Nerve Injuries; Plant Extracts; Pregabalin; Rats; Sciatic Nerve | 2022 |
The Acute Antiallodynic Effect of Tolperisone in Rat Neuropathic Pain and Evaluation of Its Mechanism of Action.
Current treatment approaches to manage neuropathic pain have a slow onset and their use is largely hampered by side-effects, thus there is a significant need for finding new medications. Tolperisone, a centrally acting muscle relaxant with a favorable side effect profile, has been reported to affect ion channels, which are targets for current first-line medications in neuropathic pain. Our aim was to explore its antinociceptive potency in rats developing neuropathic pain evoked by partial sciatic nerve ligation and the mechanisms involved. Acute oral tolperisone restores both the decreased paw pressure threshold and the elevated glutamate level in cerebrospinal fluid in neuropathic rats. These effects were comparable to those of pregabalin, a first-line medication in neuropathy. Tolperisone also inhibits release of glutamate from rat brain synaptosomes primarily by blockade of voltage-dependent sodium channels, although inhibition of calcium channels may also be involved at higher concentrations. However, pregabalin fails to affect glutamate release under our present conditions, indicating a different mechanism of action. These results lay the foundation of the avenue for repurposing tolperisone as an analgesic drug to relieve neuropathic pain. Topics: Analgesics; Animals; Disease Models, Animal; Glutamic Acid; Neuralgia; Pregabalin; Rats; Tolperisone | 2022 |
Long-Lasting Nociplastic Pain Modulation by Repeated Administration of Sigma-1 Receptor Antagonist BD1063 in Fibromyalgia-like Mouse Models.
Sigma-1 receptor (σ1R) ligands have been shown to be effective at relieving neuropathic and inflammatory pain, but have not yet been tested in experimental models of fibromyalgia. The objective of this study was to evaluate the effect of a σ1R antagonist (BD1063) compared to pregabalin. ICR-CD1 female mice were subjected to either six repeated injections of reserpine, to cause reserpine-induced myalgia (RIM6), or acidified saline intramuscular injections (ASI). In these two models, we evaluated the effect of BD1063 and pregabalin on thermal hypersensitivity, anxiety-like and depression-like behaviors, and on spinal cord gliosis. BD1063 exerted an antinociceptive effect on both reflexive (thermal hyperalgesia) and nonreflexive (anxiety- and depression-like) pain behaviors, and reduced spinal astroglial and microglial reactivity, following repeated treatment for 2 weeks. Interestingly, the effects of BD1063 were long-term, lasting several weeks after treatment discontinuation in both fibromyalgia-like models. Similar results were obtained with pregabalin, but the effects on pain behaviors lasted for a shorter length of time, and pregabalin did not significantly modulate spinal glial reactivity. The inhibitory and long-lasting effect of pharmacological blockade of σ1Rs on both sensory and affective dimensions of nociplastic-like pain and spinal cord gliosis in two experimental models of fibromyalgia support the application of this therapeutic strategy to treat fibromyalgia. Topics: Analgesics; Animals; Chronic Pain; Disease Models, Animal; Female; Fibromyalgia; Gliosis; Hyperalgesia; Mice; Mice, Inbred ICR; Pregabalin; Receptors, sigma; Reserpine; Sigma-1 Receptor | 2022 |
Amiodarone Enhances Anticonvulsive Effect of Oxcarbazepine and Pregabalin in the Mouse Maximal Electroshock Model.
Accumulating experimental studies show that antiarrhythmic and antiepileptic drugs share some molecular mechanisms of action and can interact with each other. In this study, the influence of amiodarone (a class III antiarrhythmic drug) on the antiseizure action of four second-generation antiepileptic drugs was evaluated in the maximal electroshock model in mice. Amiodarone, although ineffective in the electroconvulsive threshold test, significantly potentiated the antielectroshock activity of oxcarbazepine and pregabalin. Amiodarone, given alone or in combination with oxcarbazepine, lamotrigine, or topiramate, significantly disturbed long-term memory in the passive-avoidance task in mice. Brain concentrations of antiepileptic drugs were not affected by amiodarone. However, the brain concentration of amiodarone was significantly elevated by oxcarbazepine, topiramate, and pregabalin. Additionally, oxcarbazepine and pregabalin elevated the brain concentration of desethylamiodarone, the main metabolite of amiodarone. In conclusion, potentially beneficial action of amiodarone in epilepsy patients seems to be limited by neurotoxic effects of amiodarone. Although results of this study should still be confirmed in chronic protocols of treatment, special precautions are recommended in clinical conditions. Coadministration of amiodarone, even at low therapeutic doses, with antiepileptic drugs should be carefully monitored to exclude undesired effects related to accumulation of the antiarrhythmic drug and its main metabolite, desethylamiodarone. Topics: Amiodarone; Animals; Anticonvulsants; Avoidance Learning; Behavior, Animal; Brain; Disease Models, Animal; Electroshock; Epilepsy; Mice; Oxcarbazepine; Pregabalin | 2021 |
Wide-Range Measurement of Thermal Preference-A Novel Method for Detecting Analgesics Reducing Thermally-Evoked Pain in Mice.
Wide use of oxaliplatin as an antitumor drug is limited by severe neuropathy with pharmacoresistant cold hypersensitivity as the main symptom. Novel analgesics to attenuate cold hyperalgesia and new methods to detect drug candidates are needed.. We developed a method to study thermal preference of oxaliplatin-treated mice and assessed analgesic activity of intraperitoneal duloxetine and pregabalin used at 30 mg/kg. A prototype analgesiameter and a broad range of temperatures (0-45 °C) were used. Advanced methods of image analysis (deep learning and machine learning) enabled us to determine the effectiveness of analgesics. The loss or reversal of thermal preference of oxaliplatin-treated mice was a measure of analgesia.. Duloxetine selectively attenuated cold-induced pain at temperatures between 0 and 10 °C. Pregabalin-treated mice showed preference towards a colder plate of the two used at temperatures between 0 and 45 °C.. Unlike duloxetine, pregabalin was not selective for temperatures below thermal preferendum. It influenced pain sensation at a much wider range of temperatures applied. Therefore, for the attenuation of cold hypersensitivity duloxetine seems to be a better than pregabalin therapeutic option. We propose wide-range measurements of thermal preference as a novel method for the assessment of analgesic activity in mice. Topics: Analgesics; Animals; Antineoplastic Agents; Cold Temperature; Disease Models, Animal; Duloxetine Hydrochloride; Hot Temperature; Hyperalgesia; Male; Mice; Oxaliplatin; Pain; Pain Measurement; Pregabalin; Temperature | 2021 |
Pregabalin-induced neuroprotection and gait improvement in dystrophic MDX mice.
Duchenne muscular dystrophy (DMD) is a genetic disease linked to the X chromosome induced by mutations in the dystrophin gene. Neuroprotective drugs, such as pregabalin (PGB), can improve motor function through the modulation of excitatory synapses, together with anti-apoptotic and anti-inflammatory effects. The present work studied the effects of PGB in the preservation of dystrophic peripheral nerves, allowing motor improvements in MDX mice. Five weeks old MDX and C57BL/10 mice were treated with PGB (30 mg/kg/day, i.p.) or vehicle, for 28 consecutive days. The mice were sacrificed on the 9th week, the sciatic nerves were dissected out and processed for immunohistochemistry and qRT-PCR, for evaluating the expression of proteins and gene transcripts related to neuronal activity and Schwann cell function. The lumbar spinal cords were also processed for qRT-PCR to evaluate the expression of neurotrophic factors and pro- and anti-inflammatory cytokines. Cranial tibial muscles were dissected out for endplate evaluation with α-bungarotoxin. The recovery of motor function was monitored throughout the treatment, using a spontaneous walking track test (Catwalk system) and a forced locomotion test (Rotarod). The results showed that treatment with PGB reduced the retrograde effects of muscle degeneration/regeneration on the nervous system from the 5th to the 9th week in MDX mice. Thus, PGB induced protein expression in neurons and Schwann cells, protecting myelinated fibers. In turn, better axonal morphology and close-to-normal motor endplates were observed. Indeed, such effects resulted in improved motor coordination of dystrophic animals. We believe that treatment with PGB improved the balance between excitatory and inhibitory inputs to spinal motoneurons, increasing motor control. In addition, PGB enhanced peripheral nerve homeostasis, by positively affecting Schwann cells. In general, the present results indicate that pregabalin is effective in protecting the PNS during the development of DMD, improving motor coordination, indicating possible translation to the clinic. Topics: Animals; Disease Models, Animal; Gait; Male; Mice; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; Neuroprotection; Neuroprotective Agents; Pregabalin; Recovery of Function; Sciatic Nerve | 2021 |
Interactions among Lacosamide and Second-Generation Antiepileptic Drugs in the Tonic-Clonic Seizure Model in Mice.
Combination therapy with two or three antiseizure medications (ASMs) is sometimes a preferred method of treatment in epilepsy patients. (1) Background: To detect the most beneficial combination among three ASMs, a screen test evaluating in vivo interactions with respect to their anticonvulsant properties, was conducted on albino Swiss mice; (2) Methods: Classification of interactions among lacosamide (LCM) and selected second-generation ASMs (lamotrigine (LTG), pregabalin (PGB), oxcarbazepine (OXC), and topiramate (TPM)) was based on the isobolographic analysis in the mouse maximal electroshock-induced seizure (MES) model. Interactions among LCM and second-generation ASMs were visualized using a polygonogram; (3) Results: In the mouse MES model, synergy was observed for the combinations of LCM + TPM + PGB and LCM + OXC + PGB. Additivity was reported for the other combinations tested i.e., LCM + LTG + TPM, LCM + LTG + PGB, LCM + LTG + OXC, and LCM + OXC + TPM in this seizure model. No adverse effects associated with triple ASM combinations, containing LCM and second-generation ASMs were observed in mice; (4) Conclusions: The combination of LCM + TPM + PGB was the most beneficial combination among the tested in this study, offering synergistic suppression of tonic-clonic seizures in mice subjected to the MES model. Both the isobolographic analysis and polygonogram method can be recommended for experimental epileptology when classifying interactions among the ASMs. Topics: Animals; Anticonvulsants; Disease Models, Animal; Drug Interactions; Drug Synergism; Drug Therapy, Combination; Electroshock; Epilepsy; Lacosamide; Lamotrigine; Male; Mice; Oxcarbazepine; Pregabalin; Seizures; Topiramate | 2021 |
Assessment of the Anti-Allodynic and Anti-Hyperalgesic Efficacy of a Glycine Transporter 2 Inhibitor Relative to Pregabalin, Duloxetine and Indomethacin in a Rat Model of Cisplatin-Induced Peripheral Neuropathy.
Cisplatin, which is a chemotherapy drug listed on the World Health Organisation's List of Essential Medicines, commonly induces dose-limiting side effects including chemotherapy-induced peripheral neuropathy (CIPN) that has a major negative impact on quality of life in cancer survivors. Although adjuvant drugs including anticonvulsants and antidepressants are used for the relief of CIPN, analgesia is often unsatisfactory. Herein, we used a rat model of CIPN (cisplatin) to assess the effect of a glycine transporter 2 (GlyT2) inhibitor, relative to pregabalin, duloxetine, indomethacin and vehicle. Male Sprague-Dawley rats with cisplatin-induced mechanical allodynia and mechanical hyperalgesia in the bilateral hindpaws received oral bolus doses of the GlyT2 inhibitor (3-30 mg/kg), pregabalin (3-100 mg/kg), duloxetine (3-100 mg/kg), indomethacin (1-10 mg/kg) or vehicle. The GlyT2 inhibitor alleviated both mechanical allodynia and hyperalgesia in the bilateral hindpaws at a dose of 10 mg/kg, but not at higher or lower doses. Pregabalin and indomethacin induced dose-dependent relief of mechanical allodynia but duloxetine lacked efficacy. Pregabalin and duloxetine alleviated mechanical hyperalgesia in the bilateral hindpaws while indomethacin lacked efficacy. The mechanism underpinning pain relief induced by the GlyT2 inhibitor at 10 mg/kg is likely due to increased glycinergic inhibition in the lumbar spinal cord, although the bell-shaped dose-response curve warrants further translational considerations. Topics: Analgesics; Animals; Benzamides; Cisplatin; Disease Models, Animal; Dose-Response Relationship, Drug; Duloxetine Hydrochloride; Glycine Plasma Membrane Transport Proteins; Hyperalgesia; Indomethacin; Male; Peripheral Nervous System Diseases; Pregabalin; Rats; Rats, Sprague-Dawley; Treatment Outcome | 2021 |
Investigation of the Combination of Pregabalin with Duloxetine or Amitriptyline on the Pharmacokinetics and Antiallodynic Effect During Neuropathic Pain in Rats.
Amitriptyline, duloxetine, and pregabalin are among the most pharmacotherapeutic, effective treatments for neuropathic pain control. However, the evaluation of synergism by combining these treatments is still poorly investigated.. To evaluate the pharmacokinetics of the combination of pregabalin plus duloxetine and pregabalin plus amitriptyline, as well as the effect of these on neuropathic pain on rodent model.. The experimental study.. The research took place in the research laboratories at the Federal University of Alfenas after ethics committee approval.. This study used male Wistar rats weighing between 220 and 250 g. The animals were randomly divided into the following groups: monotherapy (pregabalin, amitriptyline, duloxetine), combined therapy (pregabalin + amitriptyline, pregabalin + duloxetine), and vehicle (ultrapure water). Pharmacokinetic analysis of pregabalin or combination (pregabalin + amitriptyline or pregabalin + duloxetine) in the plasma were performed by ultraperformance liquid chromatography tandem mass spectrometry. Neuropathic pain was induced by sciatic nerve constriction (chronic constriction injury [CCI]) model, and nociceptive threshold was measured by von Frey filaments test. In addition, to evaluate the influence of the treatments on the motor coordination, the rotarod test was used.. The pharmacokinetic disposition of pregabalin was changed in the association with amitriptyline, presenting a clearance reduction and consequently an increase in bioavailability. Furthermore, after the 14th day of CCI, pregabalin was administered orally and induced antiallodynic effect after 1, 2:15, 4, and 8 hours of its administration and showed the greatest antiallodynic effect after 4 hours of its administration. Moreover, this effect was prolonged (up to 8 hours) by combination with amitriptyline. Additionally, pregabalin and pregabalin + duloxetine showed a hypoalgesic effect in sham rats. In addition, the rotarod test results showed that drugs did not influence the motor coordination of the rats.. Potential competition mechanisms during the excretion of pregabalin, when pregabalin was combined with amitriptyline, were not investigated in this study.. The data demonstrated that combined therapy of pregabalin plus amitriptyline improved the bioavailability of pregabalin and potentiated the efficacy of the antiallodynic effect of pregabalin alone, proving to be advantageous for the treatment of sciatic neuropathic pain. Topics: Amitriptyline; Analgesics; Animals; Disease Models, Animal; Duloxetine Hydrochloride; Hyperalgesia; Male; Neuralgia; Pain Measurement; Pregabalin; Rats; Rats, Wistar | 2021 |
Pregabalin Treatment does not Affect Amyloid Pathology in 5XFAD Mice.
Calcium dysregulation has been proposed to play a causative role in the development of Alzheimer's disease pathology. Pregabalin is a compound already approved for human use, marketed as the prescription drug Lyrica. It binds the α2-δ subunit of P/Q-type voltagegated calcium channels, lowering calcium influx and providing effective treatment for epilepsy and neuropathic pain.. We hypothesize that increased resting calcium in neuronal processes near amyloid plaques plays a role in the development of neuritic dystrophies and further progression of amyloid pathology.. 5XFAD mice were treated orally for 12 weeks with pregabalin, then immunoblotting and immunofluorescent imaging were used to quantify neuritic dystrophy and amyloid deposition in pregabalin compared to placebo-treated mice.. The treatment did not decrease markers of neuritic dystrophy or amyloid deposition. The image analysis of neuritic dystrophy on a plaque-by-plaque basis showed a small non-significant increase in the relative proportion of LAMP1 to Aβ42 in plaques with areas of 50-450 μm2 in the cortex of pregabalin-treated mice. In addition, there was a statistically significant positive correlation between the measured cerebral concentration of pregabalin and the relative levels of BACE1 and Aβ in the cortex. This relationship was not observed in the hippocampus, and there was no increase in average Aβ levels in pregabalin treated mice compared to placebo. We confirmed previous findings that smaller amyloid plaques are associated with a greater degree of neuritic dystrophy.. Pregabalin may have an effect on Aβ that merits further investigation, but our study does not suggest that pregabalin contributes substantially to amyloid pathology. Topics: Alzheimer Disease; Amyloid Precursor Protein Secretases; Animals; Disease Models, Animal; Hippocampus; Humans; Mice; Mice, Transgenic; Neurites; Plaque, Amyloid; Pregabalin | 2021 |
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection. Topics: Animals; Antiviral Agents; Artificial Intelligence; Chlorocebus aethiops; Disease Models, Animal; Drug Evaluation, Preclinical; High-Throughput Screening Assays; Immunocompetence; Inhibitory Concentration 50; Methacycline; Mice, Inbred C57BL; Protease Inhibitors; Quantitative Structure-Activity Relationship; Small Molecule Libraries; Vero Cells; Zika Virus; Zika Virus Infection | 2020 |
Pregabalin inhibits in vivo and in vitro cytokine secretion and attenuates spleen inflammation in Lipopolysaccharide/Concanavalin A -induced murine models of inflammation.
Immune system alteration has been implicated in the pathogenesis of chronic pain conditions, epilepsy and generalized anxiety disorder. Targeting cytokines has recently been proposed for the management of such conditions. Pregabalin (PGB) is an antiepileptic agent used for the management of these conditions. However, little is known about its immunomodulatory effects on cytokine secretion in vivo and in vitro. Hence, a mitogen (Lipopolysaccharide [LPS] or Concanavalin A [ConA])-induced murine model of inflammation was used to investigate the effect of PGB on in vivo and in vitro IL-1β, IL-6, TNF-α and IL-2 cytokine secretion using ELISA. In addition, PGB effect on spleen histology, as a lymphoid organ, was examined. Our results revealed that PGB significantly inhibited the secretion of ConA-induced IL-6 secretion, basal and ConA-induced TNF-α and IL-2 secretion in splenocytes in vitro. In vivo, PGB inhibited basal and LPS/ConA-induced IL-6 and TNF-α secretion in addition to LPS-induced IL-1β and ConA-induced IL-2 secretion. Moreover, PGB attenuated mitogen-induced inflammatory changes in the spleen. These findings provide an evidence of the anti-inflammatory properties of PGB on cytokine secretion and lymphoid organ inflammation. This might give insights into the role of PGB in the management of the inflammatory state in PGB-indicated conditions. Topics: Animals; Concanavalin A; Cytokines; Disease Models, Animal; Female; Inflammation; Lipopolysaccharides; Mice; Mice, Inbred BALB C; Pregabalin; Spleen | 2020 |
Prenatal pregabalin is associated with sex-dependent alterations in some behavioral parameters in valproic acid-induced autism in rat offspring.
This study was performed to evaluate the effects of prenatal exposure to pregabalin (PGB) on behavioral changes of rat offspring in an animal model of valproic acid (VPA)-induced autism-like symptoms. Pregnant rats received VPA (600 mg/kg/i.p.) once at 12.5 gestational days for autism-like symptom induction in offspring. After the delivery single male and single female offspring from each mother were randomly selected for behavioral test (anxiety, pain response, pleasure, and motor function) at 60th day adulthood (n = 7). Offspring received prenatal PGB (15 & 30 mg/kg/i.p.) during gestational days 9.5 to 15.5 either alone or in combination with VPA (PGB15, PGB30, PGB15 + VPA, and PGB30 + VPA). Control offspring received normal saline during the same period. The result showed that prenatal VPA exposure was associated with autism-like behaviors in rat offspring. PGB treatment during the gestational period revealed significant reduction in sucrose preference test and anxiety in elevated plus maze and open field test in offspring. Also, PGB treatments exhibited a dose-dependent increase in pain threshold in prenatally VPA exposed rats in tail-flick and hot plate test. Also, there was a sex-related significant impairment in motor function in beam balance and open field test, and male rats were affected more than females. However, no significant sex differences in sucrose preference and pain sensitivity were observed in prenatal PGB-treated rat offspring. In conclusion, prenatal exposure to VPA increased the risk of autism-like behaviors in the offspring rats, and PGB treatment during the gestational period was associated with some beneficial effects, including anxiety reduction and motor impairment in autism-like symptoms in rat offspring. Topics: Animals; Anxiety; Autistic Disorder; Behavior, Animal; Choice Behavior; Disease Models, Animal; Female; Male; Motor Activity; Pain Threshold; Pregabalin; Pregnancy; Prenatal Exposure Delayed Effects; Rats; Sex Characteristics; Sex Factors; Valproic Acid | 2020 |
Assessment of the anti-allodynic efficacy of a glycine transporter 2 inhibitor relative to pregabalin and duloxetine in a rat model of prostate cancer-induced bone pain.
The pathobiology of prostate cancer-induced bone pain (PCIBP) is underpinned by both inflammatory and neuropathic components. Here, we used a rat model of PCIBP to assess the analgesic efficacy of a glycine transporter 2 (GlyT2) inhibitor (N-(6-((1,3-dihydroxypropan-2-yl)amino)-2-(dimethylamino)pyridin-3-yl)-3,5-dimethoxy-4-(4-(trifluoromethyl)phenoxy) benzamide) relative to two clinically available adjuvant drugs that are recommended for the relief of neuropathic pain, viz, pregabalin and duloxetine.. PCIBP was induced in male Wistar Han rats following intra-tibial injection (ITI) of rat prostate cancer (AT3B) cells into the left tibia. Sham-rats received an ITI of heat-killed AT3B cells. PCIBP rats with fully developed mechanical allodynia in the ipsilateral hindpaws as assessed using von Frey filaments, received single oral (p.o.) bolus doses of the GlyT2 inhibitor (3-30 mg/kg), pregabalin (3-100 mg/kg), duloxetine (3-100 mg/kg), or vehicle. Baseline paw withdrawal thresholds (PWTs) were determined in the ipsilateral (injured side) and contralateral hindpaws immediately prior to dosing and at scheduled times for 3 h post dosing in individual animals.. Single oral bolus doses of the GlyT2 inhibitor (3-30 mg/kg) evoked partial pain relief at the doses tested in the ipsilateral hindpaws of PCIBP rats without any discernible behavioural side effects. By contrast, single oral bolus doses of pregabalin at 10-100 mg/kg evoked dose-dependent and complete alleviation of mechanical allodynia. By comparison, single oral bolus doses of duloxetine at doses up to 100 mg/kg lacked efficacy.. Oral administration of this GlyT2 inhibitor evoked partial pain relief in PCIBP rats and did not evoke central nervous system side effects in contrast to GlyT2 inhibitors reported by others. Topics: Analgesics; Animals; Bone and Bones; Cancer Pain; Disease Models, Animal; Duloxetine Hydrochloride; Glycine Plasma Membrane Transport Proteins; Hyperalgesia; Male; Neuralgia; Pain Measurement; Pain Threshold; Pregabalin; Prostatic Neoplasms; Rats; Rats, Wistar | 2020 |
Chronic Pregabalin Treatment Ameliorates Pain, but not Depressive-Like Behaviors, in a Reserpine-Induced Myalgia Model in Rats.
Anticonvulsants are often prescribed as coanalgesics for pathologies presenting chronic pain, such as chronic neuropathic pain and fibromyalgia. These pathologies are associated with a wide range of comorbidities: chronic fatigue, cognitive impairment, sleep disturbances, and mood disorders. Pregabalin, an anticonvulsant used to treat fibromyalgia syndrome, has been proven to improve pain and fatigue symptoms. However, most studies have not considered the analytic effect of this drug on comorbid depressive-like symptoms in this syndrome.. The main study objective was to examine the role of pregabalin in depressive symptomatology comorbid to chronic widespread pain using a reserpine-induced myalgia model.. A randomized, controlled, animal study.. Research and data analyses were performed at the GESADA laboratory, Department of Human Anatomy and Embryology, University of Valencia, Spain.. Forty-six Sprague-Dawley male rats were used. Acute chronic pregabalin administration was tested for depressive-like behaviors (Forced Swimming and Novelty-Suppressed Feeding Tests) and for alteration of pain thresholds (tactile allodynia, Electronic Von Frey test; and mechanical hyperalgesia, Randall and Selitto test). The same procedures were followed with duloxetine as a positive control.. Pregabalin significantly improved depressive-like behaviors in acute, but not chronic treatment, and significantly ameliorated pain thresholds.. Lack of histological and electrophysiological tests.. Pregabalin is not effective in depressive-like symptoms associated with chronic pain but might play an acute antidepressive-like role given its antinociceptive effect. Topics: Animals; Anticonvulsants; Antihypertensive Agents; Chronic Pain; Depression; Disease Models, Animal; Drug Administration Schedule; Male; Myalgia; Pain Threshold; Pregabalin; Random Allocation; Rats; Rats, Sprague-Dawley; Reserpine; Treatment Outcome | 2020 |
A New Rat Model of Thalamic Pain Produced by Administration of Cobra Venom to the Unilateral Ventral Posterolateral Nucleus.
Thalamic pain is a neuropathic pain syndrome that occurs as a result of thalamic damage. It is difficult to develop therapeutic interventions for thalamic pain because its mechanism is unclear. To better understand the pathophysiological basis of thalamic pain, we developed and characterized a new rat model of thalamic pain using a technique of microinjecting cobra venom into the ventral posterolateral nucleus (VPL) of the thalamus.. This study will establish a new thalamic pain rat model produced by administration of cobra venom to the unilateral ventral posterolateral nucleus.. This study used an experimental design in rats.. The research took place in the laboratory at the Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine.. Male Sprague-Dawley rats were subjected to the administration of cobra venom or saline into the left VPL. The development of mechanical hyperalgesia and changes in pain-related behaviors and motor function were measured after intrathalamic cobra venom microinjection using the von Frey test, video recording, and cylinder test, respectively. On postoperative days 7 to 35, both electroacupuncture and pregabalin (PGB) were administered to verify that the model reproduced the findings in humans. Moreover, the organizational and structural alterations of the thalamus were examined via transmission electron microscopy (TEM).. The threshold for mechanical stimuli in the left facial skin was significantly decreased on day 3 after thalamic pain modeling as compared with pre-venom treatment. Furthermore, the ultrastructural alterations of neurons such as indented neuronal nuclei, damaged mitochondria and endoplasmic reticulum, and dissolved surrounding tissues were observed under TEM. Moreover, electroacupuncture treatment ameliorated mechanical hyperalgesia, pain-like behaviors, and motor dysfunction, as well as restore normal structures of neurons in the thalamic pain rat model. However, no such beneficial effects were noted when PGB was administered.. The pathophysiological features were different from the present model and the patients in clinical practice (in most cases strokes, either ischemic or hemorrhagic).. The cobra venom model may provide a reasonable model for investigating the mechanism of thalamic pain and for testing therapies targeting recovery and pain after thalamic lesions.. Thalamic pain, cobra venom, electroacupuncture, pregabalin, indented neuronal nuclei, damaged mitochondria, dissolved endoplasmic reticulum, golgi body. Topics: Animals; Brain; China; Disease Models, Animal; Elapid Venoms; Electroacupuncture; Hyperalgesia; Male; Neuralgia; Pain Measurement; Pregabalin; Rats; Rats, Sprague-Dawley; Trigeminal Neuralgia; Ventral Thalamic Nuclei | 2019 |
Ongoing pain in streptozotocin model of diabetes in the rat: correlation with cutaneous cheminociception.
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 4 Topics: Analgesics; Animals; Conditioning, Psychological; Diabetes Mellitus, Experimental; Diabetic Neuropathies; Disease Models, Animal; Male; Mustard Plant; Pain; Plant Oils; Pregabalin; Rats; Rats, Wistar; Streptozocin; TRPA1 Cation Channel | 2019 |
Tropomyosin Receptor Kinase B Receptor Activation in the Locus Coeruleus Restores Impairment of Endogenous Analgesia at a Late Stage Following Nerve Injury in Rats.
A rat model of neuropathic pain at 6 weeks after spinal nerve ligation (SNL6w) exhibits both mechanical hypersensitivity and impaired noxious stimuli-induced analgesia (NSIA). Repeated treatment with antidepressants can produce antihypersensitivity and restore NSIA. To examine the involvement of a brain-derived neurotrophic factor-mediated mechanism, a tropomyosin receptor kinase B (TrkB) agonist, 7,8-dihydroxyflavone (DHF), was administered to SNL6w rats (5 mg/kg/d for 5 days). Mechanical hypersensitivity was evaluated using the von Frey filament test and paw pressure test. NSIA was examined by measuring the change in the hind paw withdrawal threshold 30 minutes after painful stimulation induced by capsaicin injection into the fore paw. Changes in the concentrations of glutamate and GABA in the locus coeruleus area were measured by in vivo microdialysis. DHF treatment did not affect mechanical hypersensitivity, although it restored NSIA by reducing GABA release in response to the fore paw capsaicin injection. DHF treatment did not alter the baseline concentration of glutamate or GABA. These findings suggest that DHF treatment restored the stimuli-response activity of the locus coeruleus without affecting the tonic activity of the locus coeruleus. The brain-derived neurotrophic factor-TkB signaling is also involved in the NSIA-restoring effect of amitriptyline. PERSPECTIVE: This article demonstrates that repeated treatment with TrkB agonist, DHF, restored endogenous analgesia. Repeated amitriptyline treatment showed similar effect via TrkB-mediated mechanisms, and the effect may be independent from the effect of antihypersensitivity. This effect of TrkB activation is promising for patients with chronic pain with impaired descending inhibition. Topics: Analgesics; Animals; Disease Models, Animal; Flavones; gamma-Aminobutyric Acid; Glutamic Acid; Hyperalgesia; Locus Coeruleus; Male; Neuralgia; Pregabalin; Random Allocation; Rats, Sprague-Dawley; Receptor, trkB; Spinal Nerves | 2019 |
Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel-induced neuropathy in rats: Role of Notch1 receptor and JAK/STAT signaling pathway.
Peripheral neuropathy is a common adverse effect observed during the use of paclitaxel (PTX) as chemotherapy. The present investigation was directed to estimate the modulatory effect of bone marrow derived mesenchymal stem cells (BM-MSCs) on pregabalin (PGB) treatment in PTX-induced peripheral neuropathy. Neuropathic pain was induced in rats by injecting PTX (2 mg/kg, i.p) 4 times every other day. Rats were then treated with PGB (30 mg/kg/day, p.o.) for 21 days with or without a single intravenous administration of BM-MSCs. At the end of experiment, behavioral and motor abnormalities were assessed. Animals were then sacrificed for measurement of total antioxidant capacity (TAC), nerve growth factor (NGF), nuclear factor kappa B p65 (NF-κB p65), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and active caspase-3 in the sciatic nerve. Moreover, protein expressions of Notch1 receptor, phosphorylated Janus kinase 2 (p-JAK2), phosphorylated signal transducer and activator of transcription 3 (p-STAT3), and phosphorylated p38 mitogen-activated protein kinase (p-p38-MAPK) were estimated. Finally, histological examinations were performed to assess severity of sciatic nerve damage and for estimation of BM-MSCs homing. Combined PGB/BM-MSCs therapy provided an additional improvement toward reducing PTX-induced oxidative stress, neuro-inflammation, and apoptotic markers. Interestingly, BM-MSCs therapy effectively prevented motor impairment observed by PGB treatment. Combined therapy also induced a significant increase in cell homing and prevented PTX-induced sciatic nerve damage in histological examination. The present study highlights a significant role for BM-MSCs in enhancing treatment potential of PGB and reducing its motor side effects when used as therapy in the management of peripheral neuropathy. Topics: Acetone; Analysis of Variance; Animals; Antineoplastic Agents, Phytogenic; Antioxidants; Caspase 3; Cell- and Tissue-Based Therapy; Disease Models, Animal; Hyperalgesia; Interleukin-6; Janus Kinases; Male; Mesenchymal Stem Cells; Motor Disorders; Nerve Growth Factor; Paclitaxel; Pregabalin; Rats; Rats, Wistar; Receptor, Notch1; Rotarod Performance Test; Sciatic Neuropathy; STAT Transcription Factors; Time Factors; Tumor Necrosis Factor-alpha | 2019 |
Distinguishing analgesic drugs from non-analgesic drugs based on brain activation in macaques with oxaliplatin-induced neuropathic pain.
The antineoplastic agent oxaliplatin is a first-line treatment for colorectal cancer. However, neuropathic pain, characterized by hypersensitivity to cold, emerges soon after treatment. In severe instances, dose reduction or curtailing treatment may be necessary. While a number of potential treatments for oxaliplatin-induced neuropathic pain have been proposed based on preclinical findings, few have demonstrated efficacy in randomized, placebo-controlled clinical studies. This failure could be related, in part, to the use of rodents as the primary preclinical species, as there are a number of distinctions in pain-related mechanisms between rodents and humans. Also, an indicator of preclinical pharmacological efficacy less subjective than behavioral endpoints that is translatable to clinical usage is lacking. Three days after oxaliplatin treatment in Macaca fascicularis, a significantly reduced response latency to cold (10 °C) water was observed, indicating cold hypersensitivity. Cold-evoked bilateral activation of the secondary somatosensory (SII) and insular (Ins) cortex was observed with functional magnetic resonance imaging. Duloxetine alleviated cold hypersensitivity and significantly attenuated activation in both SII and Ins. By contrast, neither clinically used analgesics pregabalin nor tramadol affected cold hypersensitivity and cold-evoked activation of SII and Ins. The current findings suggest that suppressing SII and Ins activation leads to antinociception, and, therefore, could be used as a non-behavioral indicator of analgesic efficacy in patients with oxaliplatin-induced neuropathic pain. Topics: Analgesics; Animals; Antineoplastic Agents; Brain; Cerebral Cortex; Cryopyrin-Associated Periodic Syndromes; Disease Models, Animal; Duloxetine Hydrochloride; Macaca fascicularis; Magnetic Resonance Imaging; Male; Neuralgia; Oxaliplatin; Pregabalin; Somatosensory Cortex; Tramadol | 2019 |
Pregabalin Ameliorates Lipopolysaccharide-Induced Pancreatic Inflammation in Aged Rats.
The aim of this study was to examine pancreatic pathology and the prophylactic effects of pregabalin in lipopolysaccharide (LPS) induced sepsis model in aged rats.. Twenty-four female, one-year-old, Wistar Albino rats were assigned to three groups; Group I (control), Group II (study group: 5mg/kg LPS intraperitoneal, single dose) and Group III(treatment group: 5mg/kg LPS+30 mg/kg oral pregabalin one hour before LPS). Animals were sacrificed by exsanguination 6 hours after LPS administration. Blood and pancreatic tissue samples were collected for biochemical, pathological, and immunohistochemical analyses.. LPS caused increases in serum amylase and lipase level but led to a reduction in glucose levels. Following histopathological analysis, numerous neutrophil leucocyte infiltrations were observed in vessels and pancreatic tissues. Increased caspase-3 expression was observed in both the endocrine and exocrine pancreas in the LPS group. Similarly, IL-6, caspase-3 (Cas-3), inducible nitric oxide synthase (iNOS), granulocyte colony-stimulating factor (G-CSF) and serum amyloid-A (SAA) expressions were increased by LPS. Pregabalin improved biochemical, histopathological, and immunohistochemical findings.. This study showed that LPS causes pathological findings in the pancreas, but pregabalin has ameliorative effects in aged rats with sepsis. Cas-3, IL-6, iNOS, G-CSF, and SAA all play pivotal roles in the pathogenesis of LPS-induced pancreatic damage. Topics: Animals; Anti-Inflammatory Agents; Caspase 3; Cytoprotection; Disease Models, Animal; Female; Granulocyte Colony-Stimulating Factor; Inflammation Mediators; Interleukin-6; Lipopolysaccharides; Nitric Oxide Synthase Type II; Pancreas; Pancreatitis; Pregabalin; Rats, Wistar; Serum Amyloid A Protein; Signal Transduction | 2019 |
Dronedarone (a multichannel blocker) enhances the anticonvulsant potency of lamotrigine, but not that of lacosamide, pregabalin and topiramate in the tonic-clonic seizure model in mice.
Topics: Animals; Anti-Arrhythmia Agents; Anticonvulsants; Disease Models, Animal; Dronedarone; Drug Synergism; Drug Therapy, Combination; Lacosamide; Lamotrigine; Male; Mice; Pregabalin; Seizures; Topiramate | 2019 |
Synergistic symptom-specific effects of ketorolac-tramadol and ketorolac-pregabalin in a rat model of peripheral neuropathy.
Although current neuropathic pain treatment guidelines do not recommend the use of nonsteroidal anti-inflammatory drugs (NSAIDs), whether NSAIDs can serve as a useful adjuvant to conventional multimodal therapy remains unclear.. The spared nerve injury (SNI) rats rapidly developed profound and long-lasting spontaneous and evoked pain behaviors, including mechanical and cold allodynia of the ipsilateral hind paw. At day 5, we first characterized the nociceptive responses to ketorolac, tramadol, pregabalin, and their combinations.. We found that tramadol and pregabalin exerted dose-dependent analgesic effects on both spontaneous and evoked behaviors. However, ketorolac alone did not suppress any behaviors regardless of the dose. Ketorolac-tramadol and ketorolac-pregabalin produced variable degrees of additive suppression of spontaneous and evoked behavioral responses. Cold allodynia was profoundly diminished after ketorolac was added to ineffective pregabalin or tramadol. Mechanical allodynia was markedly attenuated by ketorolac-pregabalin but less so by ketorolac-tramadol mixtures.. Our data demonstrated that an NSAID alone failed to relieve spontaneous or evoked pain behaviors in the rat SNI model, but when combined with a weak opioid and α-2-δ-ligand produced a profound synergistic analgesic effect on cold allodynia and discrepant efficacy for mechanical allodynia and spontaneous pain. Topics: Animals; Disease Models, Animal; Drug Synergism; Drug Therapy, Combination; Ketorolac; Male; Meloxicam; Neuralgia; Pregabalin; Rats; Rats, Sprague-Dawley; Tramadol | 2019 |
Effects of Three Anti-Seizure Drugs on Cholinergic and Metabolic Activity in Experimental Status Epilepticus.
Status epilepticus (SE) is characterized by recurrent seizure activity and can be drug- resistant. Knowledge of neuronal and metabolic activity of the brain during SE may be helpful to improve medical care. We here report the effects of three anti-seizure drugs on changes of acetylcholine energy metabolites and oxidative stress during SE.. We used the lithium-pilocarpine model in rats to induce SE and in vivo- microdialysis to monitor cholinergic and metabolic activity in the hippocampus. We measured extracellular concentrations of acetylcholine, glucose, lactate, pyruvate, glycerol and isoprostanes before and during SE, and after acute treatment with pregabalin, valproic acid, and levetiracteam.. Upon onset of SE, acetylcholine (ACh) release increased six- to eightfold. Glucose was increased only transiently by 30% but lactate levels rose four-fold, and extracellular concentrations of glycerol ten-fold. Isoprostanes are markers of oxidative stress and increased more than 20-fold. Two hours after pilocarpine adminstration, rats were treated with pregabalin (100 mg/kg), levetiracetam (200 mg/kg) or valproic acid (400 mg/kg) by i.p. injection. All three drugs stopped seizure activity in a delayed fashion, but at the doses indicated, only animals that received levetiracetam reached consciousness. All drugs reduced ACh release within 60-120 minutes. Lactate/pyruvate ratios, glycerol and isoprostanne levels were also reduced significantly after drug administration.. Hippocampal ACh release closely follows seizure activity in SE and is attenuated when SE subsides. Pregabalin, valproic acid and levetiracetam all terminate seizures in the rat SE model and attenuate cholinergic and metabolic changes within two hours. Topics: Acetylcholine; Animals; Anticonvulsants; Behavior, Animal; Cholinergic Agents; Chromatography, High Pressure Liquid; Disease Models, Animal; Levetiracetam; Male; Oxidative Stress; Pregabalin; Rats; Rats, Sprague-Dawley; Seizures; Status Epilepticus; Valproic Acid | 2019 |
Acute effect of cannabidiol on the activity of various novel antiepileptic drugs in the maximal electroshock- and 6 Hz-induced seizures in mice: Pharmacodynamic and pharmacokinetic studies.
Cannabidiol and cannabidiol-enriched products have recently attracted much attention as an add-on therapy for epilepsy, especially drug-resistant seizures. It should be, however, remembered that concomitant use of cannabidiol and antiepileptic drugs may pose a risk of interactions between them. For this reason, the aim of our study was to assess the effect of cannabidiol on the activity of selected new antiepileptic drugs in the electrically-induced seizure models in mice. We studied the effect of cannabidiol on the anticonvulsant action of topiramate, oxcarbazepine, lamotrigine, and pregabalin in the maximal electroshock-induced seizure test as well as on the activity of levetiracetam, tiagabine, lacosamide, and gabapentin in the 6 Hz seizure test in mice. We showed that cannabidiol increased the activity of topiramate, oxcarbazepine, pregabalin, tiagabine, and gabapentin. It did not affect the anticonvulsant effect of lamotrigine and lacosamide. Interestingly, cannabidiol attenuated the anticonvulsant activity of levetiracetam. Co-administration of antiepileptic drugs with cannabidiol did not cause adverse effects such as impairment of motor coordination, changes in neuromuscular strength or potentiation of the cannabidiol-induced hypolocomotion. Serum and brain levels of antiepileptic drugs and cannabidiol were determined by using HPLC in order to ascertain any pharmacokinetic contribution to the observed behavioral effects. Only interaction with levetiracetam was purely pharmacodynamic in nature because no changes in serum and brain concentration of either levetiracetam or cannabidiol were observed. Increased anticonvulsant activity of topiramate, oxcarbazepine, pregabalin, tiagabine, and gabapentin could be, at least in part, related to pharmacokinetic interactions with cannabidiol because there were changes in serum and/or brain concentrations of antiepileptic drugs and/or cannabidiol. Pharmacokinetic interactions cannot be also excluded between lacosamide and cannabidiol because cannabidiol increased brain concentration of lacosamide and lacosamide increased brain concentration of cannabidiol. Further pharmacokinetic studies are required to evaluate the type of interactions between cannabidiol and novel antiepileptic drugs. Topics: Animals; Anticonvulsants; Brain; Cannabidiol; Chromatography, High Pressure Liquid; Disease Models, Animal; Drug Interactions; Drug Resistant Epilepsy; Electric Stimulation; Gabapentin; Lacosamide; Lamotrigine; Levetiracetam; Male; Mice; Oxcarbazepine; Pregabalin; Seizures; Tiagabine; Topiramate | 2019 |
Alkylsulfanyl analogs as potent α
We identified novel (3R, 5S)-3-aminomethyl-5-methanesulfanyl hexanoic acid (5a: DS75091588) and (3R, 5S)-3-aminomethyl-5-ethanesulfanyl hexanoic acid (6a: DS18430756) as sulfur-containing γ-amino acid derivatives that were useful for the treatment of neuropathic pain. These two compounds exhibited a potent analgesic effect in animal models of both type I diabetes and type II diabetes, and good pharmacokinetics. Topics: Animals; Calcium Channels; Caproates; Diabetes Mellitus, Experimental; Diabetes Mellitus, Type 1; Diabetes Mellitus, Type 2; Disease Models, Animal; Ligands; Mice; Molecular Structure; Neuralgia; Sulfhydryl Compounds | 2018 |
Effects of S 38093, an antagonist/inverse agonist of histamine H3 receptors, in models of neuropathic pain in rats.
Histamine H3 receptors are mainly expressed on CNS neurons, particularly along the nociceptive pathways. The potential involvement of these receptors in pain processing has been suggested using H3 receptor inverse agonists.. The antinociceptive effect of S 38093, a novel inverse agonist of H3 receptors, has been evaluated in several neuropathic pain models in rat and compared with those of gabapentin and pregabalin.. While S 38093 did not change vocalization thresholds to paw pressure in healthy rats, it exhibited a significant antihyperalgesic effect in the Streptozocin-induced diabetic (STZ) neuropathy model after acute and chronic administration and, in the chronic constriction injury (CCI) model only after chronic administration, submitted to the paw-pressure test. Acute S 38093 administration at all doses tested displayed a significant cold antiallodynic effect in a model of acute or repeated administration of oxaliplatin-induced neuropathy submitted to cold tail immersion, cold allodynia being the main side effect of oxaliplatin in patients. The effect of S 38093 increased following chronic administration (i.e. twice a day during 5 days) in the CCI and STZ models except in the oxaliplatin models where its effect was already maximal from the first administration The kinetics and size of effect of S 38093 were similar to gabapentin and/or pregabalin. Finally, the antinociceptive effect of S 38093 could be partially mediated by α2 adrenoreceptors desensitization in the locus coeruleus.. These results highlight the interest of S 38093 to relieve neuropathic pain and warrant clinical trials especially in chemotherapeutic agent-induced neuropathic pain.. S 38093, a new H3 antagonist/inverse agonist, displays antiallodynic and antihyperalgesic effect in neuropathic pain, especially in oxaliplatin-induced neuropathy after chronic administration. This effect of S 38093 in neuropathic pain could be partly mediated by α2 receptors desensitization in the locus coeruleus. Topics: Amines; Analgesics; Animals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Histamine Antagonists; Hyperalgesia; Male; Neuralgia; Organoplatinum Compounds; Oxaliplatin; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley | 2018 |
Operant self-administration of pregabalin in a mouse model of neuropathic pain.
Pregabalin is a first-line agent for neuropathic pain treatment whose abuse liability remains controversial. Surprisingly, studies exploring the reinforcing properties of pregabalin in operant mouse models are missing.. We evaluated the acquisition of operant pregabalin self-administration in mice exposed to a partial sciatic nerve ligation (PSNL) or a sham operation. After surgery, mice were trained in operant boxes to intravenously self-administer pregabalin at 1.5 or 3 mg/kg/inf or saline during 10 days. Thermal and mechanical sensitivity were assessed before and after self-medication, and depressive-like behaviour was evaluated after discontinuation of the treatment.. Partial sciatic nerve ligation and sham-operated mice exposed to pregabalin at 3 mg/kg/inf showed higher active responding compared to mice exposed to saline. The differences in active responding were more robust in nerve-injured than in sham-operated mice. Self-medication at either dose of pregabalin partially inhibited thermal hypersensitivity, whereas only self-medication at 3 mg/kg/inf reduced mechanical sensitivity. Finally, a depressive-like behaviour was revealed after saline treatment in nerve-injured mice, and this emotional manifestation was abolished after pregabalin treatment at the high dose.. Pregabalin showed reinforcing effects both in PSNL and sham-operated mice and attenuated the nociceptive and emotional manifestations of neuropathic pain in mice self-administering this drug. Therefore, pregabalin self-administration was related to neuropathic pain relief, but also to reinforcing properties related to psychotropic drug effects. This study reveals the improvement in nociceptive and emotional manifestations of neuropathic pain after operant pregabalin self-medication in mice and suggests the reinforcing effects of this drug in an operant paradigm.. This study shows that mice with a nerve injury self-administer pregabalin at doses effective reducing nociceptive hypersensitivity and depressive-like behaviour associated with the neuropathic pain model. Interestingly, mice without neuropathy also develop operant self-administration behaviour, suggesting potential abuse liability of this first-line drug for neuropathic pain treatment. Topics: Analgesics; Animals; Behavior, Animal; Conditioning, Operant; Disease Models, Animal; Hyperalgesia; Male; Mice; Neuralgia; Peripheral Nerve Injuries; Pregabalin; Sciatic Nerve; Self Administration | 2018 |
Pregabalin induces conditioned place preference in the rat during the early, but not late, stage of neuropathic pain.
The present study aimed to examine the rewarding effects of pain relief during the early and late stages of neuropathic pain using a conditioned place preference (CPP) test. Animal models of neuropathic pain were prepared by spinal nerve ligation in male Sprague-Dawley rats. Intraperitoneal and intrathecal injections of pregabalin (300 mg/kg and 100 μg/10 μL, respectively) suppressed allodynia in the von Frey test both 2 weeks (early stage) and 4 weeks (late stage) after nerve injury. Intraperitoneal and intrathecal injections of pregabalin induced CPP during the early stage of neuropathic pain, suggesting that the CPP test serves as an objective and quantifiable behavioral assay to assess the emotional aspect of pain relief. In contrast with the early stage of neuropathic pain, intraperitoneal or intrathecal injection of pregabalin did not induce CPP during the late stage of neuropathic pain. The extinguishment of the rewarding effects of pregabalin during the late stage of neuropathic pain is likely due to dysfunction of the mesolimbic reward system, although the possibility that neuronal mechanisms other than dysfunction of the mesolimbic reward system are involved in the extinguishment of pregabalin-induced CPP cannot be excluded. We previously reported that not only the dopamine release in the nucleus accumbens induced by intrathecal pregabalin injection but also that induced by sucrose intake were extinguished during the late stage of neuropathic pain. These findings, combined with the results of this study, suggest that pain chronification leads to dysfunction of the mesolimbic reward system. Topics: Analgesics; Animals; Conditioning, Classical; Disease Models, Animal; Hyperalgesia; Male; Neuralgia; Pregabalin; Rats; Rats, Sprague-Dawley; Reward | 2018 |
Therapeutic effects of diclofenac, pregabalin, and duloxetine on disuse-induced chronic musculoskeletal pain in rats.
The aim of this study was to clarify the mechanism of disuse-induced muscle hyperalgesia through the evaluation of the pharmacological behaviour of muscle hyperalgesia profiles in chronic post-cast pain (CPCP) rats with acute and chronic-phase mirror-image muscle hyperalgesia treated with diclofenac (NSAID), pregabalin (an inhibitor of Ca Topics: Animals; Chronic Pain; Diclofenac; Disease Models, Animal; Duloxetine Hydrochloride; Humans; Muscle, Skeletal; Muscular Disorders, Atrophic; Musculoskeletal Pain; Pain Measurement; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley | 2018 |
Effects of pregabalin and duloxetine on neurotransmitters in the dorsal horn of the spinal cord in a rat model of fibromyalgia.
Dysfunction of the monoamine systems in the nervous system is associated with the clinical symptoms of fibromyalgia. Reserpine-induced myalgia (RIM) rats are a putative model of fibromyalgia in which muscle pressure thresholds and monoamine content is reduced in the brain and spinal cord. We examined the effects of pregabalin and duloxetine, drugs approved for fibromyalgia treatment, on the levels of extracellular neurotransmitters in the dorsal horn of the spinal cord in RIM rats using microdialysis. Male SD rats were used for all experiments. To generate RIM rats, reserpine was injected at 1 mg/kg subcutaneously once daily for three consecutive days. The pressure threshold of the mid-gastrocnemius muscle was measured using a Randall-Selitto apparatus. Norepinephrine, dopamine, and serotonin were detected using high-performance liquid chromatography with electrochemical detection, and glutamate and γ-aminobutyric acid (GABA) were detected using liquid chromatography-mass spectrometry. The muscle pressure threshold in RIM rats was significantly lower than that in normal rats. While the levels of monoamines and glutamate were lower in the spinal cord of RIM rats than in normal rats, levels of GABA did not markedly differ. Duloxetine increased the levels of all three monoamines in normal and RIM rats in a dose-dependent manner. In contrast, pregabalin only increased norepinephrine levels in RIM rats. These results indicate that while both pregabalin and duloxetine ameliorate muscle pressure thresholds in RIM rats, their effects on the levels of extracellular neurotransmitters in the spinal cord differ considerably. Topics: Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Duloxetine Hydrochloride; Fibromyalgia; Male; Neurotransmitter Agents; Pregabalin; Rats; Rats, Sprague-Dawley; Spinal Cord Dorsal Horn | 2018 |
Effects of pregabalin on brain edema, neurologic and histologic outcomes in experimental traumatic brain injury.
Brain edema and increased intracranial pressure (ICP) are among the main causes of neurological disturbance and mortality following traumatic brain injury (TBI). Since pregabalin neuroprotective effects have been shown, this study was performed to evaluate the possible neuroprotective effects of pregabalin in experimental TBI of male rats. Adult male Wistar rats were divided into 4 groups: sham, vehicle, pregabalin 30 mg/kg and pregabalin 60 mg/kg. TBI was induced in vehicle and pregabalin groups by Marmarou method. Pregabalin was administered 30 min after TBI. Sham and vehicle groups received saline. Brain water and Evans blue content and histopathological changes were evaluated 24, 5 and 24 h after TBI, respectively. The ICP and neurological outcomes (veterinary coma scale, VCS) were recorded before, 1 h and 24 h post TBI. The results showed a significant reduction in brain water content and ICP, and a significant increase in VCS of pregabalin group (60 mg/kg) as compared to vehicle group (P < 0.05). Also, pregabalin reduced brain edema and apoptosis score as compared to vehicle group. Post TBI pregabalin administration revealed a delayed but significant improvement in ICP and neurological outcomes in experimental TBI. The underlying mechanism(s) was not determined and needs further investigation. Topics: Animals; Apoptosis; Brain; Brain Edema; Brain Injuries, Traumatic; Capillary Permeability; Disease Models, Animal; Dose-Response Relationship, Drug; Intracranial Pressure; Male; Neuroprotective Agents; Pregabalin; Rats, Wistar | 2018 |
Pain-Related Behavior and Brain Activation in a Cynomolgus Macaque Model of Postoperative Pain.
Inadequate postoperative pain management could lead to persistent pain and this is, in part, due to incomplete understanding of the mechanism of postoperative pain. Currently available rodent models may have limited translatability to clinical postoperative pain. Thus, a preclinical model of postoperative pain was developed in the cynomolgus macaque, a species that is phylogenetically closer to humans than rodents.. The presence of pressure hypersensitivity was assessed with non-noxious pressure applied proximally and distally (approximately 10 cm) to an abdominal incision in macaques. The effect of the opioid morphine (intramuscular, i.m.), the nonsteroidal anti-inflammatory drug diclofenac (i.m.) and the anticonvulsant pregabalin (i.m.) on pressure hypersensitivity was evaluated one and two days following surgery. Brain activation during non-noxious pressure stimulation was observed with functional magnetic resonance imaging.. Hypersensitivity to non-noxious pressure applied proximally and distally (approximately 10 cm) to the incision was observed, lasting for up to seven days and three days, respectively, following surgery. Postoperative pressure hypersensitivity was attenuated with morphine but not with either diclofenac or pregabalin. Bilateral activation of the insular cortex and cingulate cortex was observed during non-noxious pressure stimulation proximal to the incision, which was attenuated with morphine. By contrast, pregabalin reduced only cingulate cortex activation.. The lack of antinociceptive efficacy of pregabalin on postoperative pain could be due to the incomplete suppression of pressure-evoked brain activation. It is speculated that incomplete postoperative pain relief observed in general could be due to residual or persistent activity of key pain nuclei such as the insular cortex. The current macaque model could be used for further elaborating the mechanism of postoperative pain as well as confirming the efficacy of potential treatments for the management of postoperative pain. Topics: Analgesics; Animals; Brain; Disease Models, Animal; Hyperalgesia; Image Processing, Computer-Assisted; Macaca fascicularis; Magnetic Resonance Imaging; Male; Oxygen; Pain, Postoperative; Physical Stimulation; Pregabalin; Time Factors | 2018 |
Combination of phenobarbital with phenytoin and pregabalin produces synergy in the mouse tonic-clonic seizure model: An isobolographic analysis.
Despite many antiepileptic drugs (AEDs) are available to treat epilepsy, there is still about 30% of epilepsy patients inadequately treated with these AEDs. For these patients, polytherapy with two or three AEDs to fully control their seizure attacks is recommended. Unfortunately, polytherapy is always associated with drug interactions, whose nature may be beneficial, neutral or unfavorable. To determine a type of interaction for the combination of three AEDs (i.e., phenobarbital [PB], phenytoin [PHT] and pregabalin [PGB]) at the fixed-ratio of 1:1:1, we used a model of tonic-clonic seizures in male albino Swiss mice.. Tonic-clonic seizures in mice were evoked by a current (sine-wave, 25 mA, 500 V, 0.2 s stimulus duration) delivered via auricular electrodes. The anticonvulsant effects of the three-drug combination (PB, PHT and PGB) in terms of suppression of tonic-clonic seizures in mice were assessed with type I isobolographic analysis. Potential acute side effects for the mixture of PB, PHT and PGB along with total brain concentrations of the AEDs were determined to confirm pharmacodynamic nature of observed interaction.. The three-drug combination of PB, PHT and PGB (at the fixed-ratio of 1:1:1) exerted synergistic interaction (at P < 0.01) in the mouse model of tonic-clonic seizures. The combination of PB, PHT and PGB did not produce any side effects in experimental animals, when measuring long-term memory, muscular strength and motor coordination. The measurement of total brain concentrations of PB, PHT and PGB was conducted to confirm that none of the three AEDs significantly influenced total brain concentrations (pharmacokinetic profiles) of the other co-administered AEDs in mice.. The synergistic pharmacodynamic interaction for the combination of PB, PHT and PGB observed in this preclinical study can be translated into clinical settings and this favorable AED combination is worthy of being recommended to some patients with refractory epilepsy. Topics: Animals; Anticonvulsants; Avoidance Learning; Brain; Disease Models, Animal; Drug Synergism; Drug Therapy, Combination; Electric Stimulation; Male; Mice; Muscle Strength; Phenobarbital; Phenytoin; Pregabalin; Seizures | 2018 |
Chronic pelvic pain and prostate inflammation in rat experimental autoimmune prostatitis: Effect of a single treatment with phosphodiesterase 5 inhibitors on chronic pelvic pain.
Experimental autoimmune prostatitis (EAP) is most often used as a nonbacterial model of chronic prostatitis/chronic pelvic pain. We investigated the development of chronic pelvic pain and inflammatory changes in rat EAP and examined the effect of a single treatment with phosphodiesterase 5 (PDE5) inhibitors on the chronic pelvic pain.. EAP was induced in rats by intradermal injection of rat prostate antigen and complete Freund's adjuvant on days 0 and 28. On day 42, after antigen injection, prostatic inflammatory changes, including the mRNA and protein levels of cytokines/chemokines, were measured and histological analysis of the prostate was performed. Pelvic pain was measured by applying von Frey filaments to the lower abdomen. To confirm that this model is appropriate for evaluating pelvic pain, we tested two drugs, celecoxib and pregabalin, which are clinically used for the treatment of prostatitis-related pain. Subsequently, we examined the effects of single treatments with three phosphodiesterase 5 inhibitors, including tadalafil, on pelvic pain in this model.. On day 42, after antigen injection, the mRNA levels of 44 of 84 kinds of cytokines/chemokines and their receptors increased significantly in EAP rats, as did the protein levels of seven of 23 kinds of cytokines/chemokines. Histological analysis revealed inflammation characterized by neutrophils and/or mononuclear cells in the glandular and stromal tissue of the ventral prostate from rats in the EAP group. Some animals in this group showed fibrosis and hemorrhage in the stromal tissue. Pelvic pain had developed in EAP rats, which was attenuated by a single treatment with celecoxib or pregabalin, suggesting that EAP is an appropriate model for prostatitis-related pain. A single treatment with any of the three PDE5 inhibitors tested attenuated the chronic pelvic pain.. Prostatitis leads to inflammatory changes in the prostate, which may contribute to the development and maintenance of chronic pelvic pain. PDE5 inhibitors, including tadalafil, may have the ability to block chronic pelvic pain. Topics: Analgesics; Animals; Autoimmune Diseases; Celecoxib; Chemokines; Chronic Pain; Cytokines; Disease Models, Animal; Immunoglobulin G; Male; Pelvic Pain; Phosphodiesterase 5 Inhibitors; Pregabalin; Prostatitis; Rats; Rats, Wistar; RNA, Messenger; Tadalafil | 2018 |
Time-shifted co-administration of sub-analgesic doses of ambroxol and pregabalin attenuates oxaliplatin-induced cold allodynia in mice.
Oxaliplatin-induced cold allodynia is a frequent complication appearing in patients treated with this anti-tumor drug. Since, there are no clear algorithms to overcome this painful condition effectively, it is important to establish novel strategies for its treatment.. In this study, the ability of pregabalin and ambroxol, used as single drugs or in combinations administered in a time-shifted manner to attenuate cold allodynia was assessed in the mouse cold plate test. The hot plate test was additionally used to assess antinociceptive properties of ambroxol in the acute, thermally-induced pain model. Locomotor activity and motor coordination of mice were also evaluated. In silico studies were undertaken to predict potential binding of ambroxol to sodium channel (Na. A hyperadditive antiallodynic effect of combined sub-analgesic ambroxol and pregabalin was demonstrated in oxaliplatin-treated mice. This effect was particularly strong when these drugs were given 4 h apart. Both drugs used in combination reduced animals' locomotor activity, but they did not impair motor coordination in the rotarod test. Ambroxol did not show antinociceptive properties in the hot plate test. The molecular docking studies predicted that in mice ambroxol might bind to Na. Time-shifted co-administration of sub-analgesic doses of ambroxol and pregabalin effectively attenuates oxaliplatin-induced cold allodynia. Molecular docking model predicts preferential binding of ambroxol to mouse Na Topics: Ambroxol; Analgesics; Animals; Behavior, Animal; Binding Sites; Cold Temperature; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Schedule; Drug Therapy, Combination; Hyperalgesia; Male; Mice; Molecular Docking Simulation; Motor Activity; NAV1.6 Voltage-Gated Sodium Channel; NAV1.9 Voltage-Gated Sodium Channel; Oxaliplatin; Pain Threshold; Pregabalin; Protein Binding; Rotarod Performance Test; Signal Transduction; Time Factors | 2018 |
Controlled cortical impact-induced neurodegeneration decreases after administration of the novel calpain-inhibitor Gabadur.
One aspect of secondary injury in traumatic brain injury is the marked increase in intracellular calcium and resultant over-activation of the calcium-dependent neutral cysteine protease calpain. Gabadur is a novel protease inhibitor with calpain-inhibition properties formulated from the classic protease inhibitor leupeptin linked to a pregabalin carrier. This construction allows the entire compound to cross the blood-brain barrier after peripheral administration to better target the site of injury. In this study, a single intraperitoneal dose of Gabadur was administered immediately following controlled cortical impact injury in rats. Neocortical slices were examined at 48 h post-injury via Fluoro-Jade B staining, revealing an improvement in cortical neurodegeneration in Gabadur treated rats. Levels of detrimental active calpain-2 measured via western blot were also decreased in rats receiving Gabadur. This data supports the benefit of targeted protease inhibition in the treatment of traumatic brain injury. Topics: Animals; Brain; Brain Injuries, Traumatic; Calpain; Disease Models, Animal; Glycoproteins; Leupeptins; Molecular Structure; Neurodegenerative Diseases; Neurons; Neuroprotective Agents; Pregabalin; Rats, Sprague-Dawley | 2018 |
Effects of pregabalin on neurobehavior in an adult male rat model of PTSD.
Posttraumatic stress disorder (PTSD) can be a very debilitating condition. Effective approaches to prevent and treat PTSD are important areas of basic science research. Pregabalin (PGB), a gabapentinoid derivative of γ-aminobutyric acid, possesses the potential to positively affect neurobehavioral changes associated with PTSD. Using a rodent model of PTSD, the aims of this study were to determine the effects of PGB as a possible prevention for the development of PTSD-like symptoms and its use as a possible treatment. A prospective, experimental, between groups design was used in conjunction with a three-day restraint/shock PTSD stress model. Sixty rats were randomly assigned between two groups, non-stressed and stressed (PTSD). Each of the main two groups was then randomly assigned into six experimental groups: control vehicle, control PGB, control naïve, PTSD vehicle, PTSD Pre-PGB (prophylactic), PTSD Post-PGB (non-prophylactic). The neurobehavioral components of PTSD were evaluated using the elevated plus maze (EPM), Morris water maze (MWM), and forced swim test (FST). Pregabalin administered 24 hours before the initial PTSD event or for 10 days following the last PTSD stress event did not statistically improve mean open arm exploration on the EPM, spatial memory, and learning in the MWM or behavioral despair measured by the FST (p > 0.05). Topics: Animals; Disease Models, Animal; Exploratory Behavior; Humans; Male; Maze Learning; Pregabalin; Prospective Studies; Rats; Spatial Memory; Stress Disorders, Post-Traumatic; Swimming | 2018 |
Analgesia induced by the epigenetic drug, L-acetylcarnitine, outlasts the end of treatment in mouse models of chronic inflammatory and neuropathic pain.
Background L-acetylcarnitine, a drug marketed for the treatment of chronic pain, causes analgesia by epigenetically up-regulating type-2 metabotropic glutamate (mGlu2) receptors in the spinal cord. Because the epigenetic mechanisms are typically long-lasting, we hypothesized that analgesia could outlast the duration of L-acetylcarnitine treatment in models of inflammatory and neuropathic pain. Results A seven-day treatment with L-acetylcarnitine (100 mg/kg, once a day, i.p.) produced an antiallodynic effect in the complete Freund adjuvant mouse model of chronic inflammatory pain. L-Acetylcarnitine-induced analgesia persisted for at least 14 days after drug withdrawal. In contrast, the analgesic effect of pregabalin, amitryptiline, ceftriaxone, and N-acetylcysteine disappeared seven days after drug withdrawal. L-acetylcarnitine treatment enhanced mGlu2/3 receptor protein levels in the dorsal region of the spinal cord. This effect also persisted for two weeks after drug withdrawal and was associated with increased levels of acetylated histone H3 bound to the Grm2 gene promoter in the dorsal root ganglia. A long-lasting analgesic effect of L-acetylcarnitine was also observed in mice subjected to chronic constriction injury of the sciatic nerve. In these animals, a 14-day treatment with pregabalin, amitryptiline, tramadol, or L-acetylcarnitine produced a significant antiallodynic effect, with pregabalin displaying the greatest efficacy. In mice treated with pregabalin, tramadol or L-acetylcarnitine the analgesic effect was still visible 15 days after the end of drug treatment. However, only in mice treated with L-acetylcarnitine analgesia persisted 37 days after drug withdrawal. This effect was associated with an increase in mGlu2/3 receptor protein levels in the dorsal horns of the spinal cord. Conclusions Our findings suggest that L-acetylcarnitine has the unique property to cause a long-lasting analgesic effect that might reduce relapses in patients suffering from chronic pain. Topics: Acetylcarnitine; Amitriptyline; Analgesics; Animals; Chronic Disease; Disease Models, Animal; Epigenesis, Genetic; Freund's Adjuvant; Hyperalgesia; Inflammation; Male; Mice; Mice, Inbred C57BL; Neuralgia; Pain Management; Pregabalin; Receptors, Metabotropic Glutamate; Time Factors; Tramadol | 2017 |
Levetiracetam synergizes with gabapentin, pregabalin, duloxetine and selected antioxidants in a mouse diabetic painful neuropathy model.
We have reported that levetiracetam, a novel anticonvulsant with analgesic properties, synergizes with ibuprofen/aspirin/paracetamol in a model of diabetic painful neuropathy (DPN). Most guidelines recommend gabapentin, pregabalin, and duloxetine as first- or second-line agents for DPN.. We examined the effects of combination treatment of first-/second-line analgesics with levetiracetam in a model of DPN. Additionally, the levetiracetam's combinations with antioxidants, low dose of aspirin, coenzyme Q10, or α-lipoic acid were evaluated.. Diabetes was induced in C57BL/6 mice with a single high dose of streptozotocin. The antinociceptive effects of orally administered levetiracetam, gabapentin, pregabalin, duloxetine (acute treatment) and aspirin, coenzyme Q10, and α-lipoic acid (preventive 7-day treatment), as well as combinations of levetiracetam with individual drugs were examined in the tail-flick test. In combination experiments, the drugs were coadministered in fixed-dose fractions of single-drug ED. About 60-, 32-, 30-, 26-, 18-, and 6-fold reductions of doses of both drugs in levetiracetam combinations with pregabalin, gabapentin, coenzyme Q10, aspirin, duloxetine, and α-lipoic acid, respectively, were detected.. Combinations of levetiracetam with gabapentin/pregabalin/duloxetine that target different mechanisms/sites of action involved in DPN, as well as combinations of levetiracetam and low-dose aspirin/coenzyme Q10/α-lipoic acid that target underlying causes of DPN, produce marked synergistic interactions in reducing nociception in diabetic mice. This suggests that these combination treatments might be of great benefit for diabetic patients and should be explored further in clinical trials. Topics: Amines; Analgesics; Animals; Anticonvulsants; Antioxidants; Cyclohexanecarboxylic Acids; Diabetes Mellitus, Experimental; Diabetic Neuropathies; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Duloxetine Hydrochloride; Gabapentin; gamma-Aminobutyric Acid; Levetiracetam; Male; Mice; Mice, Inbred C57BL; Piracetam; Pregabalin | 2017 |
A novel therapeutic with two SNAP-25 inactivating proteases shows long-lasting anti-hyperalgesic activity in a rat model of neuropathic pain.
Topics: Animals; Botulinum Toxins; Calcitonin Gene-Related Peptide; Capsaicin; Cells, Cultured; Disease Models, Animal; Hyperalgesia; Male; Models, Molecular; Motor Activity; Neuralgia; Peptide Hydrolases; Pregabalin; Rats; Rats, Sprague-Dawley; Recombinant Fusion Proteins; Sensory Receptor Cells; Synaptosomal-Associated Protein 25; Time Factors; Trigeminal Ganglion | 2017 |
Ivabradine attenuates the anticonvulsant potency of lamotrigine, but not that of lacosamide, pregabalin and topiramate in the tonic-clonic seizure model in mice.
Topics: Acetamides; Animals; Anticonvulsants; Benzazepines; Brain; Cardiovascular Agents; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Electroshock; Epilepsy, Tonic-Clonic; Fructose; Ivabradine; Lacosamide; Lamotrigine; Male; Mice; Pregabalin; Topiramate; Triazines | 2017 |
Risk to heroin users of polydrug use of pregabalin or gabapentin.
To examine the risk to heroin users of also using gabapentin or pregabalin (gabapentoids).. Multi-disciplinary study: we (a) examined trends in drug-related deaths and gabapentoid prescription data in England and Wales to test for evidence that any increase in deaths mentioning gabapentin or pregabalin is associated with trends in gabapentoid prescribing and is concomitant with opioid use; (b) interviewed people with a history of heroin use about their polydrug use involving gabapentin and pregabalin; and (c) studied the respiratory depressant effects of pregabalin in the absence and presence of morphine in mice to determine whether concomitant exposure increased the degree of respiratory depression observed.. England and Wales.. Interviews were conducted with 30 participants (19 males, 11 female).. (a) Office of National Statistics drug-related deaths from 1 January 2004 to 31 December 2015 that mention both an opioid and pregabalin or gabapentin; (b) subjective views on the availability, use, interactions and effects of polydrug use involving pregabalin and gabapentin; and (c) rate and depth of respiration.. Pregabalin and gabapentin prescriptions increased approximately 24% per year from 1 million in 2004 to 10.5 million in 2015. The number of deaths involving gabapentoids increased from fewer than one per year prior to 2009 to 137 in 2015; 79% of these deaths also involved opioids. The increase in deaths was correlated highly with the increase in prescribing (correlation coefficient 0.94; 5% increase in deaths per 100 000 increase in prescriptions). Heroin users described pregabalin as easy to obtain. They suggested that the combination of heroin and pregabalin reinforced the effects of heroin but were concerned it induced 'blackouts' and increased the risk of overdose. In mice, a low dose of S-pregabalin (20 mg/kg) that did not itself depress respiration reversed tolerance to morphine depression of respiration (resulting in 35% depression of respiration, P < 0.05), whereas a high dose of S-pregabalin (200 mg/kg) alone depressed respiration and this effect summated with that of morphine.. For heroin users, the combination of opioids with gabapentin or pregabalin potentially increases the risk of acute overdose death through either reversal of tolerance or an additive effect of the drugs to depress respiration. Topics: Adult; Amines; Analgesics; Animals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Drug Interactions; Drug Overdose; Drug Users; England; Female; Gabapentin; gamma-Aminobutyric Acid; Heroin Dependence; Humans; Male; Mice; Middle Aged; Pregabalin; Risk; Wales; Young Adult | 2017 |
Effect of different doses of pregabalin on skeletal muscle ischaemia-reperfusion injury in rats.
Analgesic, anti-inflammatory and anti-apoptotic effects of pregabalin have been shown previously. In this study, we investigated the protective effect of different doses of pregabalin on skeletal muscle IR injury in rats.. 24 rats were randomly divided into 4 groups (Control, Ischaemia-Reperfusion (IR), IR-Pregabalin 50 mg, IR-Pregabalin 200 mg). Following IR, serum Ischemia Modified Albumin (IMA) and tissue Paraoxonase (PON) were studied and gastrocnemius muscle tissue was removed for histopathologic examination.. Interstitial inflammation was higher in the IR group than in the control and Pregabalin 200 mg groups (p = 0.037, p = 0.037, respectively). Congestion was higher in the IR group than in the control, Pregabalin 50 and 200 mg groups (p = 0.001, p = 0.004, p = 0.004, respectively). PON was lower in the IR group than in the Control, Pregabalin 50 and 200 mg groups (p = 0.001, p = 0.007, p = 0.015, respectively). IMA was higher in the IR group than in the Control, Pregabalin 50 and 200 mg groups (p < 0.0001, all).. We think that administration of pregabalin, more prominent at 200 mg, can reverse the injury that occurs in the skeletal muscle of IR-induced rats. Pregabalin can be safely used for analgesia in cases of IR (Tab. 2, Fig. 9, Ref. 41). Topics: Animals; Apoptosis; Biomarkers; Disease Models, Animal; Male; Muscle, Skeletal; Oxidative Stress; Pregabalin; Rats; Rats, Wistar; Reperfusion Injury; Serum Albumin; Serum Albumin, Human | 2017 |
Therapeutic potential for leukocyte elastase in chronic pain states harboring a neuropathic component.
Neuropathic pain is an integral component of several chronic pain conditions and poses a major health problem worldwide. Despite emerging understanding of mechanisms behind neuropathic pain, the available treatment options are still limited in efficacy or associated with side effects, therefore making it necessary to find viable alternatives. In a genetic screen, we recently identified SerpinA3N, a serine protease inhibitor secreted in response to nerve damage by the dorsal root ganglion neurons and we showed that SerpinA3N acts against induction of neuropathic pain by inhibiting the T-cell- and neutrophil-derived protease, leucocyte elastase (LE). In the current study, via detailed in vivo pharmacology combined with analyses of evoked- and spontaneous pain-related behaviors in mice, we report that on systemic delivery, a single dose of 3 independent LE inhibitors can block established nociceptive hypersensitivity in early and late phases in the spared nerve injury model of traumatic neuropathic pain in mice. We further report the strong efficacy of systemic LE inhibitors in reversing ongoing pain in 2 other clinically relevant mouse models-painful diabetic neuropathy and cancer pain. Detailed immunohistochemical analyses on the peripheral tissue samples revealed that both T-Lymphocytes and neutrophils are the sources of LE on peripheral nerve injury, whereas neutrophils are the primary source of LE in diabetic neuropathic conditions. In summary, our results provide compelling evidence for a strong therapeutic potential of generic LE inhibitors for the treatment of neuropathic pain and other chronic pain conditions harboring a neuropathic pain component. Topics: Analgesics; Animals; Cancer Pain; Chronic Pain; Cyclic S-Oxides; Disease Models, Animal; Female; Glycine; Hyperalgesia; Leukocyte Elastase; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Motor Activity; Neuralgia; Pain Measurement; Pain Threshold; Pregabalin; Serine Proteinase Inhibitors; Sulfonamides; Thiazoles | 2017 |
A rat model of FOLFOX-induced neuropathy: effects of oral dimiracetam in comparison with duloxetine and pregabalin.
The FOLFOX family of chemotherapy regimens are hampered by the development of a painful neuropathy. Current clinical treatments are inadequate, and furthermore, the research of innovative drugs is strongly disadvantaged by the absence of a preclinical model based on the complete mixture of FOLFOX components. The aim of this study was to set up a rat model of FOLFOX-induced neuropathy in rats, validate its predictability by reference drugs, and evaluate the effectiveness of the new anti-neuropathic compound dimiracetam.. Male Sprague-Dawley rats were treated intraperitoneally with the FOLFOX components (6 mg kg. FOLFOX reduced the pain threshold in response to mechanical noxious and thermal (cold) non-noxious stimuli beginning from day 14 up to day 42 comparably to oxaliplatin alone. A fifth FOLFOX injection enhanced the severity but not the duration of painful alterations. Spontaneous activity, behavioural, autonomic, and neurological functions were also affected, whereas the motor coordination was not altered. On day 22, duloxetine (15 mg kg. A clinically consistent model of FOLFOX-induced neurotoxicity has been developed in rats. Dimiracetam fully reduced hypersensitivity and neurological alterations showing a relevant profile as anti-neuropathic resource. Topics: Administration, Oral; Animals; Disease Models, Animal; Duloxetine Hydrochloride; Fluorouracil; Humans; Imidazoles; Neuralgia; Pregabalin; Pyrroles; Rats; Rats, Sprague-Dawley | 2017 |
Neuroprotective effects of pregabalin in a rat model of intracisternal facial nerve avulsion.
Pregabalin (PGB), a drug used for treating neuropathic pain, has immune-modulating property that may have therapeutic implications. Suppression of microglial activation and improvement in functional recovery was observed in experimental spinal cord injury after PGB administration. An experimental study was conducted to evaluate whether PGB could afford neuroprotection in a rat model of intracisternal facial nerve avulsion.. Twenty-eight male Wistar rats (250-300 g) were dichotomized into two groups: a PGB group (N.=14) and a control group (N.=14). The PGB group received a total of 4 intraperitoneal PGB injections (30 mg/kg, 15 minutes preoperatively and 4, 24, and 48 hours postoperatively), and the control group underwent intraperitoneal saline injection. Intracisternal facial nerve avulsion was created by tangential pull-out of the nerve surgically exposed at the stylomastoid foramen. In both groups, the brainstem containing the facial motor nuclei neurons was thin-sliced and stained with cresyl violet, and the number of viable neurons in the facial motor nuclei on days 14 and 28 was counted under microscope.. The total viable neuron count was significantly greater in the PGB group than in the Control group both on day 14 (271.4±14.9 vs. 196.2±22.2, P<0.01) and day 28 (160.2±21.6 vs. 102.6±13.4, P<0.01). Furthermore, CD11b/c immunostaining on days 3 and 8 showed that CD11b/c-positive cells, suggestive of activated microglia, were observed only in the control group.. Better neuronal survival by PGB administration may be beneficial and clinically relevant when surgical reconstruction of the facial nerve, such as hypoglossal-facial nerve anastomosis, is considered. Topics: Animals; Brain Stem; Disease Models, Animal; Facial Nerve Injuries; Male; Motor Neurons; Neuroprotective Agents; Pregabalin; Rats; Rats, Wistar | 2017 |
Effects of pregabalin on spinal d-serine content and NMDA receptor-mediated synaptic transmission in mice with neuropathic pain.
Pregabalin (PGB) is a chemical derivative of the inhibitory neurotransmitter γ-aminobutyric acid, and is successfully used for the treatment of neuropathic pain. Substantial evidence suggests that d-serine, an endogenous co-agonist at the strychnine-insensitive glycine site of the NMDA receptor, counteracts the antinociceptive actions of PGB at the level of the spinal cord. In the present study, we examined the impact of PGB treatment on spinal d-serine content and NMDA receptor-mediated synaptic transmission in the superficial dorsal horn of peripheral nerve-ligated neuropathic mice. Mechanical allodynia was assessed using von Frey filaments. On post-surgical day 9 (after 5days of treatment with PGB [50mg/kg] or saline vehicle), the lumbar spinal cord was removed, homogenized, and ultrafiltrated. Supernatant samples were treated with Marfey's reagent and analyzed with liquid chromatography-mass spectrometry to measure d-serine content. In the electrophysiological experiments, tight-seal whole-cell recording was performed on neurons in the superficial dorsal horn of spinal cord slices. Partial sciatic nerve ligation increased spinal d-serine content, increased the NMDA/non-NMDA ratio of EPSC amplitudes, and slowed the decay phase of the NMDA component of EPSCs (NMDA-EPSCs). PGB treatment attenuated mechanical allodynia and reduced spinal d-serine content, decreased the NMDA/non-NMDA ratio, and shortened the decay time of NMDA-EPSCs. Furthermore, bath-applied d-serine attenuated the effects of PGB treatment. Although the precise mechanism for the effect of PGB on d-serine metabolism and abundance is unknown, the antinociceptive action of PGB likely involves the reduction of spinal d-serine content and subsequent attenuation of NMDA receptor-mediated synaptic transmission in the superficial dorsal horn. Topics: Animals; Disease Models, Animal; Excitatory Postsynaptic Potentials; gamma-Aminobutyric Acid; Male; Mice; Neuralgia; Neurons; Pregabalin; Receptors, N-Methyl-D-Aspartate; Serine; Synaptic Transmission | 2017 |
Effect of pregabalin on fear-based conditioned avoidance learning and spatial learning in a mouse model of scopolamine-induced amnesia.
Cognitive deficits are one of the frequent symptoms accompanying epilepsy or its treatment.. In this study, the effect on cognition of intraperitoneally administered antiepileptic drug, pregabalin (10 mg/kg), was investigated in scopolamine-induced memory-impaired mice in the passive avoidance task and Morris water maze task. The effect of scopolamine and pregabalin on animals' locomotor activity was also studied.. In the retention phase of the passive avoidance task, pregabalin reversed memory deficits induced by scopolamine (p < 0.05). During the acquisition phase of the Morris water maze pregabalin-treated memory-impaired mice performed the test with longer escape latencies than the vehicle-treated mice (significant at p < 0.05 on Day 5, and at p < 0.001 on Day 6). There were no differences in this parameter between the scopolamine-treated control group and pregabalin-treated memory-impaired mice, which indicated that pregabalin had no influence on spatial learning in this task. During the probe trial a significant difference (p < 0.05) was observed in terms of the mean number of target crossings between vehicle-treated mice and pregabalin-treated memory-impaired mice but there was no difference between the scopolamine-treated control group and mice treated with pregabalin + scopolamine. Pregabalin did not influence locomotor activity increased by scopolamine.. In passive avoidance task, pregabalin reversed learning deficits induced by scopolamine. In the Morris water maze, pregabalin did not influence spatial learning deficits induced by scopolamine. These results are relevant for epileptic patients treated with pregabalin and those who use it for other therapeutic indications (anxiety, pain). Topics: Amnesia; Animals; Anticonvulsants; Avoidance Learning; Conditioning, Psychological; Disease Models, Animal; Fear; Male; Maze Learning; Mice; Mice, Inbred C57BL; Pregabalin; Scopolamine; Spatial Learning; Species Specificity | 2017 |
Effect of pregabalin administration upon reperfusion in a rat model of hyperglycemic stroke: Mechanistic insights associated with high-mobility group box 1.
Hyperglycemia, which reduces the efficacy of treatments and worsens clinical outcomes, is common in stroke. Ability of pregabalin to reduce neuroexcitotoxicity may provide protection against stroke, even under hyperglycemia. We investigated its protective effect against hyperglycemic stroke and its possible molecular mechanisms. Male Wistar rats administered dextrose to cause hyperglycemia, underwent middle cerebral artery occlusion for 1 h and subsequent reperfusion. Rats were treated with an intraperitoneal injection of 30 mg/kg pregabalin or an equal amount of normal saline at the onset of reperfusion (n = 16 per group). At 24 h after reperfusion, neurological deficit, infarct volume, and apoptotic cell count were assessed. Western blot analysis was performed to determine protein expression of high-mobility group box 1 (HMGB1), toll-like receptor-4 (TLR-4), phosphorylated nuclear factor-kappa B (p-NF-κB), interleukin-1beta (IL-1β), tumor necrosis factor-alpha (TNF-α), phosphorylated inducible and endothelial nitric oxide synthase (p-iNOS, p-eNOS), Bcl-2, Bax, Cytochrome C, and caspase-3 in the brain. Pregabalin-treated rats showed significantly improved neurological function (31% decrease in score), reduced infarct size (by 33%), fewer apoptotic cells (by 63%), and lower expression levels of HMGB1, TLR4, p-NF-κB, IL-1β, and TNF- α, compared with control rats. Decreased p-iNOS and increased p-eNOS expressions were also observed. Expression of Bax, Cytochrome C, and cleaved caspase-3/caspase3 was significantly downregulated, while Bcl-2 expression was increased by pregabalin treatment. Pregabalin administration upon reperfusion decreased neuronal death and improved neurological function in hyperglycemic stroke rats. Cogent mechanisms would include attenuation of HMGB1/TLR-4-mediated inflammation and favorable modulation of the NOS. Topics: Animals; Apoptosis; Brain; Brain Infarction; Cytokines; Disease Models, Animal; HMGB1 Protein; Hyperglycemia; Injections, Intraperitoneal; Male; Neuroprotective Agents; NF-kappa B; Nitric Oxide Synthase Type II; Nitric Oxide Synthase Type III; Pregabalin; Rats; Rats, Wistar; Reperfusion Injury; Stroke; Toll-Like Receptor 4 | 2017 |
Isobolographic Analysis of Drug Combinations With Intrathecal BRL52537 (κ-Opioid Agonist), Pregabalin (Calcium Channel Modulator), AF 353 (P2X3 Receptor Antagonist), and A804598 (P2X7 Receptor Antagonist) in Neuropathic Rats.
Neuropathic pain should be treated with drug combinations exhibiting multiple analgesic mechanisms of action because the mechanism of neuropathic pain involves multiple physiological causes and is mediated by multiple pathways. In this study, we defined the pharmacological interaction of BRL52537 (κ-opioid agonist), pregabalin (calcium channel modulator), AF 353 (P2X3 receptor antagonist), and A804598 (P2X7 receptor antagonist).. Animal models of neuropathic pain were established by spinal nerve ligation (SNL) in male Sprague-Dawley rats, and responses to the mechanical stimulation using von Frey filaments were measured. Drugs were administered by intrathecal route and were examined for antiallodynic effects, and drug interactions were evaluated using isobolographic analysis. The mRNA expression levels of pain-related receptors in each spinal cord or dorsal root ganglion of naïve, SNL, and drug-treated SNL rats were evaluated using real-time polymerase chain reaction.. Intrathecal BRL52537, pregabalin, AF 353, and A804598 produced antiallodynic effects in SNL rats. In the drug combination studies, intrathecal coadministration of BRL52537 with pregabalin or A804598 exhibited synergistic interactions, and other drugs combinations showed additivity. The rank order of potency was observed as follows: BRL52537 + pregabalin > BRL52537 + A804598 > pregabalin + AF 353 > A804598 + pregabalin > BRL52537 + AF 353 > AF 353 + A804598. Real-time polymerase chain reaction indicated that alterations of P2X3 receptor and calcium channel mRNA expression levels were observed, while P2X7 receptor and κ-opioid receptor expression levels were not altered.. These results demonstrated that intrathecal combination of BRL52537, pregabalin, AF 353, and A804598 synergistically or additively attenuated allodynia evoked by SNL, which suggests the possibility to improve the efficacy of single-drug administration. Topics: Analgesics; Animals; Behavior, Animal; Calcium; Calcium Channels; Disease Models, Animal; Drug Combinations; Guanidines; Hyperalgesia; Male; Neuralgia; Pain Threshold; Piperidines; Pregabalin; Purinergic P2X Receptor Antagonists; Pyrrolidines; Quinolines; Rats; Rats, Sprague-Dawley | 2017 |
Synergistic anticonvulsant effects of pregabalin and amlodipine on acute seizure model of epilepsy in mice.
Status epilepticus is a life threatening neurological medical emergency. It may cause serious damage to the brain and even death in many cases if not treated properly. There is limited choice of drugs for the short term and long term management of status epilepticus and the dugs recommended for status epilepticus possess various side effects. The present study was designed to investigate synergistic anticonvulsant effects of pregabalin with amlodipine on acute seizure model of epilepsy in mice. Pentylenetetrazole was used to induce acute seizures which mimic status epilepticus. Pregabalin and amlodipine were used in combination to evaluate synergistic anti-seizure effects on acute seizure model of epilepsy in mice. Diazepam and valproate were used as reference dugs. The acute anti-convulsive activity of pregabalin with amlodipine was evaluated in vivo by the chemical induced seizures and their anti-seizure effects were compared with pentylenetetrazole, reference drugs and to their individual effects. The anti-seizure effects of tested drugs were recorded in seconds on seizure characteristics such as latency of onset of threshold seizures, rearing and fallings and Hind limbs tonic extensions. The seizure protection and mortality to the animals exhibited by the drugs were recorded in percentage. Combination regimen of pregabalin with amlodipine exhibited dose dependent significant synergistic anticonvulsant effects on acute seizures which were superior to their individual effects and equivalent to reference drugs. Topics: Amlodipine; Animals; Anticonvulsants; Disease Models, Animal; Drug Synergism; Drug Therapy, Combination; Epilepsy; Male; Mice; Pentylenetetrazole; Pregabalin; Seizures | 2017 |
Development of Novel Alkoxyisoxazoles as Sigma-1 Receptor Antagonists with Antinociceptive Efficacy.
A novel series of sigma (σ) receptor ligands based on an alkoxyisoxazole scaffold has been designed and synthesized. Preliminary receptor binding assays identified highly potent (Ki < 1 nM) and selective σ1 ligands devoid of binding interactions with the monoamine transporters DAT, NET, and SERT. In particular, compound 53 was shown to possess significant antinociceptive activity in the mouse formalin-induced inflammation pain model when administered intraperitoneally at 40 and 80 mg/kg. Initial pharmacokinetics evaluation indicated an excellent brain exposure following oral dosing in mice, suggesting that further investigation into the use of alkoxyisoxazoles as σ1 ligands for antinociception is warranted. This study supports the notion that selective σ1 antagonism could be a useful strategy in the development of novel antipain therapy. Topics: Analgesics; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Formaldehyde; Injections, Intraperitoneal; Isoxazoles; Ligands; Mice; Mice, Inbred Strains; Molecular Structure; Pain; Receptors, sigma; Sigma-1 Receptor; Structure-Activity Relationship | 2016 |
Influence of MPEP (a selective mGluR5 antagonist) on the anticonvulsant action of novel antiepileptic drugs against maximal electroshock-induced seizures in mice.
The aim of this study was to determine the effects of 2-methyl-6-(phenylethynyl)pyridine (MPEP - a selective antagonist for the glutamate metabotropic receptor subtype mGluR5) on the protective action of some novel antiepileptic drugs (lamotrigine, oxcarbazepine, pregabalin and topiramate) against maximal electroshock-induced seizures in mice. Brain concentrations of antiepileptic drugs were measured to determine whether MPEP altered pharmacokinetics of antiepileptic drugs. Intraperitoneal injection of 1.5 and 2mg/kg of MPEP significantly elevated the threshold for electroconvulsions in mice, whereas MPEP at a dose of 1mg/kg considerably enhanced the anticonvulsant activity of pregabalin and topiramate, but not that of lamotrigine or oxcarbazepine in the maximal electroshock-induced seizures in mice. Pharmacokinetic results revealed that MPEP (1mg/kg) did not alter total brain concentrations of pregabalin and topiramate, and the observed effect in the mouse maximal electroshock seizure model was pharmacodynamic in nature. Collectively, our preclinical data suggest that MPEP may be a safe and beneficial adjunct to the therapeutic effects of antiepileptic drugs in human patients. Topics: Animals; Anticonvulsants; Brain; Carbamazepine; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Evaluation, Preclinical; Drug Therapy, Combination; Electroshock; Excitatory Amino Acid Antagonists; Fructose; Lamotrigine; Male; Mice; Oxcarbazepine; Pregabalin; Pyridines; Random Allocation; Receptor, Metabotropic Glutamate 5; Seizures; Topiramate; Triazines | 2016 |
Effects of pregabalin on the nociceptive, emotional and cognitive manifestations of neuropathic pain in mice.
Preclinical drug discovery for the treatment of chronic pain is at present challenged by the difficulty to study behaviours comparable to the complex human pain experience in animals. Several reports have demonstrated a frequent association of chronic pain in humans with affective disorders, such as anxiety and depression, and impaired cognitive functions, including memory and decision making, and motivation for goal-directed behaviours. In this study, we validated different behavioural outcomes to measure the emotional and cognitive manifestations of neuropathic pain induced in mice by partial sciatic nerve ligation.. In these mice, we evaluated at different time points the nociceptive responses, the anxiety- and depressive-like behaviours, the anhedonic state, object recognition memory and the operant responding maintained by food and the effects of the repeated administration of pregabalin on these manifestations.. Our results demonstrated that the presence of allodynia and hyperalgesia in neuropathic pain mice was associated with increased anxiety- and depressive-like behaviours, reduced memory functions, development of an anhedonic state and impaired motivation to obtain food in the operant task. Chronic pregabalin treatment improved the nociceptive, anxiety-like and anhedonic responses, as well as the memory deficit, but did not modify the depressive-like alterations and the decreased motivation in these mice.. These results indicate that some emotional manifestations of chronic pain do not necessarily resolve when pain is relieved and underline the relevance to evaluate multiple behavioural responses associated with chronic pain, including the affective-motivational and cognitive behaviours, to increase the predictive value of preclinical drug discovery. WHAT DOES THIS STUDY ADD?: In this study, we have validated different behavioural outcomes allowing a reliable measurement of the emotional and cognitive manifestations of neuropathic pain induced in mice by partial sciatic nerve ligation. These results underline the relevance to evaluate these multiple pain-related alterations to improve the predictive value of preclinical drug discovery. Topics: Animals; Cognition; Disease Models, Animal; Emotions; Hyperalgesia; Male; Mice; Neuralgia; Pain Measurement; Pregabalin | 2016 |
Amitriptyline, but Not Pregabalin, Reverses the Attenuation of Noxious Stimulus-Induced Analgesia After Nerve Injury in Rats.
Noxious stimulus-induced analgesia (NSIA) is a type of conditioned pain modulation in rats that has been used to assess endogenous pain control systems. The descending noradrenergic system is involved in NSIA, and nerve injury induces plastic changes of descending noradrenergic neurons. Thus, we hypothesized that nerve injury would affect NSIA strength and that amitriptyline and pregabalin, which often are used for treating neuropathic pain, might further modulate NSIA through effects on the descending noradrenergic system.. We examined the change in NSIA over time after right L5 spinal nerve ligation (SNL) in rats by measuring the contralateral hind paw withdrawal threshold after left forepaw capsaicin injection. In addition, we examined NSIA after 5 daily intraperitoneal injection of amitriptyline or pregabalin. Microdialysis studies were performed to measure noradrenaline levels after left forepaw capsaicin injection in the left spinal dorsal horn in noninjured rats, SNL rats, and SNL rats that had received 5 daily intraperitoneal injections of amitriptyline or pregabalin.. NSIA was dramatically attenuated 5 and 6 weeks after SNL (P < 0.001). The noradrenaline level in the lumbar spinal cord was significantly increased in noninjured rats receiving forepaw injection of capsaicin compared with vehicle injection (P < 0.001), but not in rats 6 weeks after SNL surgery. Five daily intraperitoneal injections of amitriptyline (10 mg/kg/d) or pregabalin (10 mg/kg/d) at 5 weeks after SNL gradually increased the ipsilateral hindpaw withdrawal threshold (P < 0.001). At 6 weeks after SNL, amitriptyline, but not pregabalin, reversed the attenuation of NSIA by SNL (P < 0.001) and increased the spinal noradrenaline level after forepaw injection of capsaicin (P = 0.005).. These data suggest that endogenous analgesia in neuropathic pain states is strongly decreased from a certain time after nerve injury and that amitriptyline reverses the attenuation of endogenous analgesia through effects on the descending noradrenergic system. Topics: Adrenergic Neurons; Amitriptyline; Analgesics; Animals; Behavior, Animal; Capsaicin; Disease Models, Animal; Microdialysis; Neuralgia; Norepinephrine; Pain Threshold; Peripheral Nerve Injuries; Pregabalin; Rats; Rats, Sprague-Dawley; Reaction Time; Sensory System Agents; Spinal Nerves; Time Factors | 2016 |
Neuronal hyperexcitability in the ventral posterior thalamus of neuropathic rats: modality selective effects of pregabalin.
Neuropathic pain represents a substantial clinical challenge; understanding the underlying neural mechanisms and back-translation of therapeutics could aid targeting of treatments more effectively. The ventral posterior thalamus (VP) is the major termination site for the spinothalamic tract and relays nociceptive activity to the somatosensory cortex; however, under neuropathic conditions, it is unclear how hyperexcitability of spinal neurons converges onto thalamic relays. This study aimed to identify neural substrates of hypersensitivity and the influence of pregabalin on central processing. In vivo electrophysiology was performed to record from VP wide dynamic range (WDR) and nociceptive-specific (NS) neurons in anesthetized spinal nerve-ligated (SNL), sham-operated, and naive rats. In neuropathic rats, WDR neurons had elevated evoked responses to low- and high-intensity punctate mechanical stimuli, dynamic brushing, and innocuous and noxious cooling, but less so to heat stimulation, of the receptive field. NS neurons in SNL rats also displayed increased responses to noxious punctate mechanical stimulation, dynamic brushing, noxious cooling, and noxious heat. Additionally, WDR, but not NS, neurons in SNL rats exhibited substantially higher rates of spontaneous firing, which may correlate with ongoing pain. The ratio of WDR-to-NS neurons was comparable between SNL and naive/sham groups, suggesting relatively few NS neurons gain sensitivity to low-intensity stimuli leading to a "WDR phenotype." After neuropathy was induced, the proportion of cold-sensitive WDR and NS neurons increased, supporting the suggestion that changes in frequency-dependent firing and population coding underlie cold hypersensitivity. In SNL rats, pregabalin inhibited mechanical and heat responses but not cold-evoked or elevated spontaneous activity. Topics: Action Potentials; Analgesics; Animals; Disease Models, Animal; Ligation; Male; Microelectrodes; Neuralgia; Neurons; Physical Stimulation; Pregabalin; Rats, Sprague-Dawley; Spinal Nerves; Thalamus | 2016 |
The influence of thiamin on the efficacy of pregabalin in rats with spinal nerve ligation (SNL)-induced neuropathic pain.
The thiamin is often used in the treatment of neuropathy, and pregabalin is often used to treat neuropathic pain. Our study examined the influence of thiamin on the efficacy of pregabalin in a rat model of spinal nerve ligation (SNL)-induced neuropathic pain.. Sprague-Dawley male rats were randomly divided into six groups. The neuropathic pain-relieving properties were measured by plantar test, cold plate test, and hot plate test after administration of pregabalin (i.v) and/or thiamin (i.p) in SNL rats 14 days after operation.. In the therapy period, pregabalin, or thiamin alone all produced antinociceptive effects in rats with neuropathic pain. And combination treatment of thiamin and pregabalin resulted in an enhanced pain relief compared to the administration of pregabalin or thiamin alone.. Combination of thiamin and pregabalin produces an additive antinociceptive effect in neuropathic pain rats, this drug combination may offer a beneficial treatment option for neuropathic pain. Topics: Analgesics; Analysis of Variance; Animals; Disease Models, Animal; Drug Administration Schedule; Drug Therapy, Combination; Hyperalgesia; Ligation; Male; Neuralgia; Pain Measurement; Pain Threshold; Physical Stimulation; Pregabalin; Rats; Rats, Sprague-Dawley; Spinal Nerves; Thiamine; Time Factors; Vitamin B Complex | 2016 |
Selective Cathepsin S Inhibition with MIV-247 Attenuates Mechanical Allodynia and Enhances the Antiallodynic Effects of Gabapentin and Pregabalin in a Mouse Model of Neuropathic Pain.
Cathepsin S inhibitors attenuate mechanical allodynia in preclinical neuropathic pain models. The current study evaluated the effects when combining the selective cathepsin S inhibitor MIV-247 with gabapentin or pregabalin in a mouse model of neuropathic pain. Mice were rendered neuropathic by partial sciatic nerve ligation. MIV-247, gabapentin, or pregabalin were administered alone or in combination via oral gavage. Mechanical allodynia was assessed using von Frey hairs. Neurobehavioral side effects were evaluated by assessing beam walking. MIV-247, gabapentin, and pregabalin concentrations in various tissues were measured. Oral administration of MIV-247 (100-200 µmol/kg) dose-dependently attenuated mechanical allodynia by up to approximately 50% reversal when given as a single dose or when given twice daily for 5 days. No behavioral deficits were observed at any dose of MIV-247 tested. Gabapentin (58-350 µmol/kg) and pregabalin (63-377 µmol/kg) also inhibited mechanical allodynia with virtually complete reversal at the highest doses tested. The minimum effective dose of MIV-247 (100 µmol/kg) in combination with the minimum effective dose of pregabalin (75 µmol/kg) or gabapentin (146 µmol/kg) resulted in enhanced antiallodynic efficacy without augmenting side effects. A subeffective dose of MIV-247 (50 µmol/kg) in combination with a subeffective dose of pregabalin (38 µmol/kg) or gabapentin (73 µmol/kg) also resulted in substantial efficacy. Plasma levels of MIV-247, gabapentin, and pregabalin were similar when given in combination as to when given alone. Cathepsin S inhibition with MIV-247 exerts significant antiallodynic efficacy alone, and also enhances the effect of gabapentin and pregabalin without increasing side effects or inducing pharmacokinetic interactions. Topics: Amines; Animals; Behavior, Animal; Cathepsins; Cyclohexanecarboxylic Acids; Dipeptides; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Gabapentin; gamma-Aminobutyric Acid; Humans; Hyperalgesia; Male; Mice; Neuralgia; Pregabalin; Protease Inhibitors | 2016 |
Comparative effects of α2δ-1 ligands in mouse models of colonic hypersensitivity.
To investigate anti-hypersensitive effects of α2δ-1 ligands in non-inflammatory and inflammation-associated colonic hypersensitivity (CHS) mouse models.. To induce an inflammation-associated CHS, 1% dextran sulfate sodium (DSS) was administered to C57Bl/6J male mice, in drinking water, for 14 d. Regarding the non-inflammatory neonatal maternal separation (NMS) -induced CHS model, wild-type C57BI/6J pups were isolated from their mother from day 2 to day 14 (P2 to P14), three hours per day (from 9:00 a.m. to 12:00 p.m.). Colorectal distension was performed by inflating distension probe from 20 μL to 100 μL by 20 μL increment step every 10 s. After a first colorectal distension (CRD), drugs were administered subcutaneously, in a cumulative manner, (Gabapentin at 30 mg/kg and 100 mg/kg; Pregabalin at 10 mg/kg and 30 mg/kg; Carbamazepine at 10 mg/kg and 30 mg/kg) and a second CRD was performed one hour after each injection.. The visceromotor response (VMR) to CRD was increased by our NMS paradigm protocol in comparison to non-handled (NH) mice, considering the highest distension volumes (80 μL: 0.783 ± 0.056 mV/s vs 0.531 ± 0.034 mV/s, P < 0.05 and 100 μL: 1.087 ± 0.056 mV/s vs 0.634 ± 0.038 mV/s, P < 0.05 for NMS and NH mice, respectively). In the inflammation-associated CHS, DSS-treated mice showed a dramatic and significant increase in VMR at 60 and 80 μL distension volumes when compared to control mice (60 μL: 0.920 ± 0.079 mV/s vs 0.426 ± 0.100 mV/s P < 0.05 and 80 μL: 1.193 ± 0.097 mV/s vs 0.681 ± 0.094 mV/s P < 0.05 for DSS- and Water-treated mice, respectively). Carbamazepine failed to significantly reduce CHS in both models. Gabapentin significantly reduced CHS in the DSS-induced model for both subcutaneous injections at 30 or 100 mg/kg. Pregabalin significantly reduced VMR to CRD in the non-inflammatory NMS-induced CHS model for the acute subcutaneous administration of the highest cumulative dose (30 mg/kg) and significantly reduced CHS in low-dose DSS-treated mice in a dose-dependent manner. Finally, the percent decrease of AUC induced by acute GBP or Pregabalin treatment were higher in the inflammatory DSS-induced CHS model in comparison to the non-inflammatory NMS-induced CHS model.. This preclinical study demonstrates α2δ-1 ligands efficacy on inflammation-associated CHS, highlighting their potential clinical interest in patients with chronic abdominal pain and moderate intestinal inflammation. Topics: Amines; Animals; Calcium Channels; Cyclohexanecarboxylic Acids; Dextran Sulfate; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Inflammatory Bowel Diseases; Ligands; Male; Mice; Mice, Inbred C57BL; Pregabalin | 2016 |
Pregabalin alleviates the nitroglycerin-induced hyperalgesia in rats.
The association between the clinical use of nitroglycerin (NTG) and migraine suggests NTG as an animal model trigger for migraine. NTG-induced hyperalgesia in rats has been extensively used as a migraine model for pre-clinical research. Pregabalin is an anti-epileptic drug and may play a role in the preventive treatment of migraine; however, the mechanism of this action remains to be clarified. Herein, we performed the present study to investigate the effect of pregabalin on the NTG-induced hyperalgesia in rats. Sixty male Sprague-Dawley rats were divided equally into six groups. Thirty minutes before NTG injection, the rats were pretreated with pregabalin. von Frey hair testing was employed to evaluate tactile sensitivity. Enzyme-linked immunosorbent assay was used to analyze plasma calcitonin gene-related peptide (CGRP) levels in the jugular vein. Immunohistochemistry was applied to detect c-Fos-immunoreactive neurons and western blot was performed to detect c-Fos protein expression in trigeminal nucleus caudalis (TNC). We found that pregabalin pretreatment alleviated the NTG-induced hyperalgesia. Moreover, pregabalin suppressed peripheral CGRP release, c-Fos-immunoreactive neurons and the protein expression of c-Fos in TNC as well. These data suggest that pregabalin could alleviate the NTG-induced hyperalgesia. Further studies are required to determine the mechanisms of action for this effect. Topics: Analgesics; Analysis of Variance; Animals; Calcitonin Gene-Related Peptide; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; Hyperalgesia; Male; Nitroglycerin; Pain Measurement; Pregabalin; Proto-Oncogene Proteins c-fos; Rats; Rats, Sprague-Dawley; Vasodilator Agents | 2015 |
A hydro-ethanolic extract of Synedrella nodiflora (L.) Gaertn ameliorates hyperalgesia and allodynia in vincristine-induced neuropathic pain in rats.
The hydro-ethanolic extract of Synedrella nodiflora (L.) Gaertn whole plant has demonstrated analgesic effects in acute pain models. The extract has also demonstrated anticonvulsant effects in murine models of experimental epilepsy. The present study illustrates an evaluation of the hydro-ethanolic extract of the plant for possible analgesic properties in hyperalgesia and allodynia associated with vincristine-induced neuropathy in rats.. Neuropathic pain was induced in Sprague-Dawley rats by injecting 100 μg/kg of vincristine sulphate on alternative days for 6 days (days 0, 2, 4, 8, 10 and 12). Vincristine-induced cold allodynia, mechanical hyperalgesia and thermal hyperalgesia were measured pre-vincristine administration and on days 15, 17 and 19 post-vincristine administration. The rats were then treated with S. nodiflora extract (SNE) (100, 300 and 1000 mg/kg), pregabalin (10, 30 and 100 mg/kg) and distilled water as vehicle daily for 5 days and pain thresholds were measured on alternate days for 3 days.. SNE and pregabalin produced analgesic properties observed as increased paw withdrawal latencies to mechanical, tactile, cold water stimuli and thermal hyperalgesic tests during the 5 days of treatment.. The findings suggest that hydro-ethanolic extract of S. nodiflora possesses anti-hyperalgesic and anti-allodynic effects in vincristine-induced neuropathic pain in rats. Topics: Analgesics; Animals; Asteraceae; Disease Models, Animal; Dose-Response Relationship, Drug; Hyperalgesia; Male; Mice; Neuralgia; Pain Threshold; Plant Extracts; Pregabalin; Rats; Rats, Sprague-Dawley; Vincristine | 2015 |
Alterations in Spinal Cord Metabolism during Treatment of Neuropathic Pain.
Therapeutic options for neuropathic pain have improved over the last 20 years yet still only provide partial relief with numerous side effects. Recently, metabolomics revealed that the concentration of the endogenous metabolite N,N-dimethylsphingosine (DMS) is increased in the spinal cord in a model of neuropathic pain. Additionally, it was shown that introduction of DMS to the central nervous system (CNS) resulted in mechanical allodynia. Here, we have examined two compounds; pregabalin (Lyrica®), a drug used to treat neuropathic pain, and N-oleoylethanolamine (NOE), an endogenous endocannabinoid-like compound that is known to affect multiple lipid pathways. We found that the concentration of DMS in the spinal cord was not significantly altered upon pregabalin treatment of rats suffering from neuropathic pain. We further explored whether modulating lipid metabolism may impact neuropathic pain by testing NOE as a potential novel therapeutic. Topics: Analgesics; Animals; Cannabinoid Receptor Agonists; Disease Models, Animal; Endocannabinoids; Ethanolamines; Male; Metabolomics; Neuralgia; Oleic Acids; Pregabalin; Rats; Rats, Sprague-Dawley; Sphingosine; Spinal Cord | 2015 |
Does Serum Visfatin Represent a Biochemical Marker to an Experimental Peripheral Neuropathic Pain in Mice.
The effects of pregabalin on neuropathic pain relief and the serum visfatin level were assessed using an experimental model of neuropathy in a study conducted on 40 male mice with sciatic nerve constriction. The mice were randomly assigned to 4 groups, each with 10 mice. The mice were subjected to experimental chronic partial constriction of the sciatic nerve and compared to sham-operated, saline-treated control mice (group I). The experimental groups (II-IV) were subjected to partial constriction of the left sciatic nerve. A series of behavioral tests, electrophysiological studies and biochemical measures were performed after 3 weeks of daily oral treatment with pregabalin (20 and 40 mg/kg in groups III and IV, respectively). The study revealed the actions of pregabalin against the nociceptive effects of chronic sciatic nerve constriction in mice (p < 0.01), including replenishment of the glutathione level (p < 0.05) and reduction of the serum visfatin level. No significant effect on the tissue malondialdehyde level was found for any of the pregabalin doses. The percentage differences in the maximum tetanic force between the ipsilateral and contra lateral legs were significant in both pregabalin-treated groups (p < 0.05). We concluded that pregabalin reduced the serum visfatin level and produced a dose-dependent antinociceptive antioxidant effect. Topics: Analgesics; Animals; Behavior, Animal; Biomarkers; Cytokines; Disease Models, Animal; Dose-Response Relationship, Drug; Electrophysiological Phenomena; Glutathione; Hyperalgesia; Male; Malondialdehyde; Mice; Muscle, Skeletal; Nicotinamide Phosphoribosyltransferase; Pregabalin; Sciatica | 2015 |
Antinociceptive effects of mirtazapine, pregabalin, and gabapentin after chronic constriction injury of the infraorbital nerve in rats.
To clarify the antiallodynic effects of the α2-adrenergic receptor antagonist mirtazapine compared with those of gabapentin and pregabalin in a rat model of orofacial neuropathic pain.. Mirtazapine (10, 30, and 100 μg), gabapentin (10, 30, and 100 μg), and pregabalin (3, 10, and 30 μg) were administered intrathecally to eight male Sprague-Dawley rats with orofacial neuropathic pain induced by chronic constriction injury of the infraorbital nerve that had been carried out 2 weeks previously. Stimulation using von Frey filaments (1.0 to 15.0 g) applied to skin innervated by the injured infraorbital nerve enabled the measurement of mechanical thresholds 0 to 180 minutes after drug injection. Time-course data for the dose-response effects were analyzed using two-way analysis of variance and the posthoc Tukey-Kramer multiple-comparison test.. Intrathecal administration of not only gabapentin and pregabalin but also mirtazapine reversed the lowered mechanical nociceptive thresholds produced by the nerve injury. The ED50 (95% confidence interval) was (in μg) 49.00 (39.71-58.29) for mirtazapine, 54.84 (46.12-63.56) for gabapentin, and 13.47 (11.24-15.69) for pregabalin.. Intraspinal administration of either mirtazapine, gabapentin, or pregabalin reverses the lowered facial mechanical thresholds produced in a rat model of trigeminal neuropathic pain. Topics: Adrenergic alpha-Antagonists; Amines; Analgesics; Animals; Cranial Nerve Injuries; Cyclohexanecarboxylic Acids; Disease Models, Animal; Dose-Response Relationship, Drug; Facial Pain; Gabapentin; gamma-Aminobutyric Acid; Injections, Spinal; Male; Mianserin; Mirtazapine; Neuralgia; Nociceptive Pain; Orbit; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Time Factors; Touch | 2014 |
The effects of Phα1β, a spider toxin, calcium channel blocker, in a mouse fibromyalgia model.
This study investigated the effects of Phα1β, pregabalin and diclofenac using an animal model of fibromyalgia (FM). Repeated administration of reserpine (0.25 mg/kg sc) once daily for three consecutive days significantly decreased thermal hyperalgesia, mechanical allodynia, and dopamine and serotonin content in the brain on the 4th day. Phα1β and pregabalin treatment completely reverted the mechanical allodynia and thermal hyperalgesia induced by reserpine treatment on the 4th day, but diclofenac was ineffective. Reserpine treatment significantly increased the immobility time in the forced swim test, which is indicative of depression in the animals. Phα1β, but not pregabalin, reduced the immobility time (56%), suggesting that Phα1β may control persistent pathological pain in FM. Topics: Animals; Brain; Calcium Channel Blockers; Diclofenac; Disease Models, Animal; Dopamine; Fibromyalgia; gamma-Aminobutyric Acid; Hyperalgesia; Male; Mice; Pain; Pregabalin; Reserpine; Serotonin; Spider Venoms; Spiders | 2014 |
Anticonvulsant activity of pregabalin in the maximal electroshock-induced seizure assay in α2δ1 (R217A) and α2δ2 (R279A) mouse mutants.
Pregabalin has been shown to have anticonvulsant, analgesic, and anxiolytic activity in animal models. Pregabalin binds with high affinity to the α2δ1 and α2δ2 subunits of voltage-gated calcium channels. In order to better understand the relative contribution that binding to either the α2δ1 or α2δ2 subunits confers on the anticonvulsant activity of pregabalin, we characterized the anticonvulsant activity of pregabalin in different wild-type (WT) and mutant mouse strains. Two targeted mouse mutants have been made in which either the α2δ1 subunit was mutated (arginine-to-alanine mutation at amino acid 217; R217A) or the α2δ2 subunit was mutated (arginine-to-alanine mutation at amino acid 279; R279A). These mutations in α2δ1 or α2δ2 render the subunits relatively insensitive to pregabalin binding. The anticonvulsant activity of pregabalin was assessed in these different mouse lines using the maximal electroshock-induced seizure (MES) model. Pregabalin reduced the percentage of seizures and increased the latency to seizure in the MES model in two parental mouse strains used to construct the mutants. Pregabalin also reduced the percentage of seizures and increased latency to seizure similarly in the α2δ2 (R279A) and WT littermate control mice. In contrast, pregabalin's anticonvulsant efficacy was significantly reduced in α2δ1 (R217A) mutants compared with WT littermate control mice. Phenytoin showed anticonvulsant activity across all WT and mutant mice. These data show that the anticonvulsant activity of pregabalin in the MES model requires binding to the α2δ1 subunit. Topics: Animals; Anticonvulsants; Calcium Channels; Disease Models, Animal; Dose-Response Relationship, Drug; Electroshock; gamma-Aminobutyric Acid; Mice, 129 Strain; Mice, Inbred C57BL; Mice, Transgenic; Mutation, Missense; Phenytoin; Pregabalin; Seizures; Species Specificity; Time Factors | 2014 |
Face-to-face comparison of the predictive validity of two models of neuropathic pain in the rat: analgesic activity of pregabalin, tramadol and duloxetine.
We compared the preclinical analgesic activity of three marketed drugs with different pharmacological properties, pregabalin, tramadol and duloxetine, described as effective against neuropathic pain in the clinic. These drugs were tested against evoked pain in two different neuropathic models in the rat, the Bennett (CCI) and the Chung (SNL) models. The selected endpoints were tactile allodynia, tactile hyperalgesia, heat hyperalgesia and cold allodynia. Although all three drugs displayed analgesic activity, the effects observed varied according to the behavioral evaluation. Pregabalin showed clear analgesic effects against cold allodynia and tactile hyperalgesia in both the CCI and Chung models. Tramadol was active against all four endpoints in the Chung model with similar effects in the CCI model, apart from tactile allodynia. Duloxetine inhibited tactile allodynia and heat hyperalgesia in both neuropathic pain models. It also displayed efficacy against tactile hyperalgesia in the CCI model and against cold allodynia in the Chung model. These data confirm that the CCI and the Chung models of neuropathic pain do not detect the activity of analgesics with the same sensitivity. Furthermore, the mode of stimulation (tactile or thermal) and the type of endpoint (allodynia or hyperalgesia) can further influence the observed efficacy of gold standards as well as novel compounds developed for treating neuropathic pain symptoms. Topics: Analgesics; Animals; Cold Temperature; Disease Models, Animal; Duloxetine Hydrochloride; gamma-Aminobutyric Acid; Hot Temperature; Hyperalgesia; Male; Neuralgia; Physical Stimulation; Pregabalin; Rats, Wistar; Sciatic Nerve; Thiophenes; Tramadol | 2014 |
Silicon-containing GABA derivatives, silagaba compounds, as orally effective agents for treating neuropathic pain without central-nervous-system-related side effects.
Neuropathic pain is a chronic condition resulting from neuronal damage. Pregabalin, the (S)-isomer of 3-isobutyl-γ-aminobutyric acid (GABA), is widely used to treat neuropathic pain, despite the occurrence of central nervous system (CNS)-related side effects such as dizziness and somnolence. Here we describe the pharmacology of novel GABA derivatives containing silicon-carbon bonds, silagaba compounds. Silagaba131, 132, and 161 showed pregabalin-like analgesic activities in animal models of neuropathic pain, but in contrast to pregabalin they did not impair neuromuscular coordination in rotarod tests. Pharmacokinetic studies showed that brain exposure to silagaba compounds was lower than that to pregabalin. Surprisingly, despite their potent analgesic action in vivo, silagaba compounds showed only weak binding to α2-δ protein. These compounds may be useful to study mechanisms of neuropathic pain. Our results also indicate that silagaba132 and 161 are candidates for orally effective treatment of neuropathic pain without CNS-related side effects. Topics: Administration, Oral; Analgesics; Animals; Brain; Disease Models, Animal; Dose-Response Relationship, Drug; gamma-Aminobutyric Acid; Hyperalgesia; Male; Mice; Neuralgia; Organosilicon Compounds; Pain Threshold; Pregabalin; Psychomotor Performance; Radioligand Assay; Rats, Sprague-Dawley; Rats, Wistar; Rotarod Performance Test; Touch | 2014 |
Novel use of perineural pregabalin infusion for analgesia in a rat neuropathic pain model.
The anticonvulsant drugs pregabalin and gabapentin are often used systemically to treat some forms of chronic neuropathic pain. However, many patients report side effects serious enough to cause discontinuation of the drug. Here we present evidence that pregabalin may block neuropathic pain when applied to the site of nerve injury in a rat neuropathic pain model.. Forty male Sprague Dawley rats were randomized into 4 groups: sciatic nerve crush injury with perineural pregabalin treatment (treatment), crush injury with perineural saline treatment (saline control), crush injury with subcutaneous pregabalin treatment (systemic drug control), and sham surgery (sham surgery control). Animals received either continuous infusions of 1% pregabalin for 7 days (treatment and systemic control) or saline (saline control) and were tested for pain behaviors using incapacitance meter, guarding scores, and radiant heat withdrawal latency (Hargreaves method). Nerves were studied using histology and immunohistochemistry for α(2)δ-1 receptors thought to mediate the central analgesic action of pregabalin.. Treatment rats had significantly better guarding scores than systemic drug controls or saline controls (P < 0.0001) and had significantly better incapacitance scores than systemic drug controls and saline controls (P ≤ 0.001). Hargreaves method data showed hypoalgesia in all injured animals with no difference among injured groups (P = 0.80). Qualitatively, immunohistochemistry likely showed equivalent expression of the α(2)δ-1 calcium channel at the injured nerve site in all nerve-injured animals.. Perineural pregabalin administration produced superior analgesia compared with that of systemic pregabalin in this neuropathic pain model. Perineural pregabalin treatment may provide a useful alternative to systemic pregabalin treatment for neuropathic pain. Topics: Analgesics; Animals; Behavior, Animal; Calcium Channels; Disease Models, Animal; gamma-Aminobutyric Acid; Infusions, Parenteral; Male; Nerve Crush; Neuralgia; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Sciatic Nerve; Sciatic Neuropathy; Time Factors | 2014 |
Effects of D-series resolvins on behavioral and neurochemical changes in a fibromyalgia-like model in mice.
This study investigated whether the spinal or systemic treatment with the lipid resolution mediators resolvin D1 (RvD1), aspirin-triggered resolvin D1 (AT-RvD1) and resolvin D2 (RvD2) might interfere with behavioral and neurochemical changes in the mouse fibromyalgia-like model induced by reserpine. Acute administration of AT-RvD1 and RvD2 produced a significant inhibition of mechanical allodynia and thermal sensitization in reserpine-treated mice, whereas RvD1 was devoid of effects. A similar antinociceptive effect was obtained by acutely treating animals with the reference drug pregabalin. Noteworthy, the repeated administration of AT-RvD1 and RvD2 also prevented the depressive-like behavior in reserpine-treated animals, according to assessment of immobility time, although the chronic administration of pregabalin failed to affect this parameter. The induction of fibromyalgia by reserpine triggered a marked decrease of dopamine and serotonin (5-HT) levels, as examined in total brain, spinal cord, cortex and thalamus. Reserpine also elicited a reduction of glutamate levels in total brain, and a significant increase in the spinal cord and thalamus. Chronic treatment with RvD2 prevented 5-HT reduction in total brain, and reversed the glutamate increases in total brain and spinal cord. Otherwise, AT-RvD1 led to a recovery of dopamine levels in cortex, and 5-HT in thalamus, whilst it diminished brain glutamate contents. Concerning pregabalin, this drug prevented dopamine reduction in total brain, and inhibited glutamate increase in brain and spinal cord of reserpine-treated animals. Our data provide novel evidence, showing the ability of D-series resolvins AT-RvD1, and mainly RvD2, in reducing painful and depressive symptoms allied to fibromyalgia in mice. Topics: Analgesics; Animals; Antidepressive Agents; Brain; Depression; Disease Models, Animal; Docosahexaenoic Acids; Dopamine; Fibromyalgia; gamma-Aminobutyric Acid; Glutamic Acid; Hot Temperature; Hyperalgesia; Male; Mice; Nociceptive Pain; Pregabalin; Serotonin; Spinal Cord; Touch | 2014 |
Inhibition of mitogen-activated protein kinases phosphorylation plays an important role in the anti-nociceptive effect of pregabalin in zymosan-induced inflammatory pain model.
Although pregabalin has been shown to have preclinical and clinical efficacy in neuropathic pain, the mechanism of its antinociceptive action is still unknown in other pain states. This study aimed to evaluate the antinociceptive effect of pregabalin and its underlying spinal mechanisms related to mitogen activated protein kinases (MAPKs) in neuron and microglia following intraplantar injection of zymosan model. Zymosan evoked thermal hyperalgesia, mechanical hyperalgesia, and mechanical allodynia starting from 1 h and persistent until 5 h post-injection, which were dose-dependently reversed by oral pretreatment of pregabalin (3, 10, and 30 mg/kg). Pregabalin dramatically inhibited zymosan-induced Fos expression (a marker for neuronal activation) and microglia activation (using markers CD11b and ED1) in the spinal dorsal horn. Moreover, zymosan significantly increased phosphorylation of extracellular signal-regulated protein kinase (ERK) 1/2 (double labeling with neuron), ERK5 (double labelling with neuron and microglia) and p38 MAPK (double labeling with microglia) in the spinal dorsal horn, which overall elevations were reversed by pregabalin. These findings suggest that blockage of MAPKs activation in neuron and microglia might be closely related to the antinociceptive effect of pregabalin on zymosan-induced peripheral inflammatory pain. Topics: Analgesics; Animals; Disease Models, Animal; gamma-Aminobutyric Acid; Male; Mitogen-Activated Protein Kinases; Pain; Pain Measurement; Phosphorylation; Pregabalin; Rats; Rats, Sprague-Dawley; Treatment Outcome; Zymosan | 2014 |
Pregabalin role in inhibition of morphine analgesic tolerance and physical dependency in rats.
Pregabalin is recently proposed as analgesic or adjuvant in pain management. While previous preclinical investigations have evaluated pregabalin-opioid interactions, the effect of pregabalin on opioid tolerance and dependency has not yet been studied. Here we evaluated the effects of different doses of pregabalin (50, 100 and 200mg/kg, s.c.) on morphine-induced tolerance and dependency in rats. Adult male Wistar rats were rendered tolerant to analgesic effect of morphine by injection of morphine (10mg/kg, s.c.) twice daily for 7 days. To develop morphine dependence, rats were given escalating doses of morphine. To determine the effect of pregabalin on the development of morphine tolerance and dependence, different doses of pregabalin were administrated before morphine. The tail-flick and naloxone precipitation withdrawal tests were used to evaluate the degree of tolerance and dependence, respectively. Chronic morphine-injected rats showed significant decrements in the percentage maximum possible effect (%MPE) of morphine on the days 5 and 7 (32.5%±3.5, 21.5%±4, respectively) compared to the first day (100%) which showed morphine tolerance. Pregabalin 200mg/kg completely prevented the development of morphine tolerance. In addition, concomitant treatment of morphine with pregabalin attenuated almost all of the naloxone-induced withdrawal signs which include weight loss, jumping, penis licking, teeth chattering, wet dog shakes, rearing, standing, sniffing, face grooming and paw tremor. These data show that pregabaline has a potential anti-tolerant/anti-dependence property against chronic usage of morphine. Therefore, pregabalin appears to be a promising candidate for the treatment of opioid addiction after confirming by future clinical studies. Topics: Analgesics, Opioid; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Tolerance; gamma-Aminobutyric Acid; Male; Morphine; Morphine Dependence; Pain Measurement; Pregabalin; Rats; Rats, Wistar; Substance Withdrawal Syndrome | 2014 |
Pregabalin reduces acute inflammatory and persistent pain associated with nerve injury and cancer in rat models of orofacial pain.
To assess the analgesic effect of pregabalin in orofacial models of acute inflammatory pain and of persistent pain associated with nerve injury and cancer, and so determine its effectiveness in controlling orofacial pains having different underlying mechanisms.. Orofacial capsaicin and formalin tests were employed in male Wistar rats to assess the influence of pregabalin (or vehicle) pretreatment in acute pain models, and the results from these experiments were analyzed by one-way analysis of variance (ANOVA) followed by Newman Keuls post-hoc test. Pregabalin (or vehicle) treatment was also tested on the facial heat hyperalgesia that was evaluated in rats receiving injection of the inflammatory irritant carrageenan into the upper lip, as well as after constriction of the infraorbital nerve (a model of trigeminal neuropathic pain), or after inoculation of tumor cells into the facial vibrissal pad; two-way repeated measures ANOVA followed by Newman-Keuls post-hoc test was used to analyze data from these experiments.. Facial grooming induced by capsaicin was abolished by pretreatment with pregabalin at 10 and 30 mg/kg. However, pregabalin failed to modify the first phase of the formalin response, but reduced the second phase at both doses (10 and 30 mg/kg). In addition, treatment of rats with pregabalin reduced the heat hyperalgesia induced by carrageenan, as well as by nerve injury and facial cancer.. Pregabalin produced a marked antinociceptive effect in rat models of facial inflammatory pain as well as in facial neuropathic and cancer pain models, suggesting that it may represent an important agent for the clinical control of orofacial pain. Topics: Acute Pain; Analgesics; Animals; Anti-Inflammatory Agents; Capsaicin; Carrageenan; Chronic Pain; Disease Models, Animal; Facial Neoplasms; Facial Pain; gamma-Aminobutyric Acid; Hot Temperature; Hyperalgesia; Irritants; Lip Diseases; Male; Neoplasm Transplantation; Orbit; Pain Measurement; Pregabalin; Random Allocation; Rats, Wistar; Sensory System Agents; Trigeminal Neuralgia | 2014 |
Synthesis and pharmacological evaluation of novel N-aryl-3,4-dihydro-1'H-spiro[chromene-2,4'-piperidine]-1'-carboxamides as TRPM8 antagonists.
A novel series of N-aryl-3,4-dihydro-1'H-spiro[chromene-2,4'-piperidine]-1'-carboxamides was identified as transient receptor potential melastatin 8 (TRPM8) channel blockers through analogue-based rational design, synthesis and screening. Details of the synthesis, effect of aryl groups and their substituents on in-vitro potency were studied. The effects of selected functional groups on the 4-position of the chromene ring were also studied, which showed interesting results. The 4-hydroxy derivatives showed excellent potency and selectivity. Optical resolution and screening of alcohols revealed that (R)-(-)-isomers were in general more potent than the corresponding (S)-(+)-isomers. The isomer (R)-(-)-10e (IC50: 8.9nM) showed a good pharmacokinetic profile upon oral dosing at 10mg/kg in Sprague-Dawley (SD) rats. The compound (R)-(-)-10e also showed excellent efficacy in relevant rodent models of neuropathic pain. Topics: Administration, Oral; Amides; Analgesics; Animals; Disease Models, Animal; Half-Life; Male; Mice; Mice, Inbred C57BL; Neuralgia; Piperidines; Protein Binding; Rats; Rats, Sprague-Dawley; Spiro Compounds; Stereoisomerism; Structure-Activity Relationship; TRPM Cation Channels | 2013 |
The antinociceptive effect of reversible monoamine oxidase-A inhibitors in a mouse neuropathic pain model.
Neuropathic pain is a debilitating condition that is often resistant to common analgesics, such as opioids, but is sensitive to some antidepressants, an effect that seems to be mediated by spinal cord 5-HT3 receptors. Because the analgesic potential of monoamine oxidase-A (MAO-A) inhibitors is understudied, we evaluated the potential antinociceptive effect of the reversible MAO-A inhibitors moclobemide and 2-(3,4-dimethoxy-phenyl)-4,5-dihydro-1H-imidazole (2-DMPI) in a mouse neuropathic pain model induced by chronic constriction injury (CCI) of the sciatic nerve. Neuropathic mice showed a decreased mechanical paw withdrawal threshold (PWT) 7 days after lesion compared with the baseline PWT, characterizing the development of hyperalgesia. Moclobemide (100-300 μmol/kg, s.c.) and 2-DMPI (30-300 μmol/kg, s.c.) treatments were able to reverse the CCI-induced hyperalgesia, with 50% inhibitory dose (ID50) values of 39 (18-84) and 11 (4-33) μmol/kg, and maximum inhibition (Imax) values of 88±14 and 98±15%, respectively, at the 300 μmol/kg dose. In addition, we observed a significant increase in the MAO-A activity in the lumbar spinal cord of CCI-submitted mice compared with sham-operated animals. Furthermore, the antihyperalgesic effects of both 2-DMPI and moclobemide were largely reversed by intrathecal injection of the 5-HT3 receptor antagonist ondansetron (10 μg/site). These results suggest a possible involvement of MAO-A in the mechanisms of neuropathic pain and a potential utility of the reversible inhibitors of MAO-A in the development of new therapeutic approaches to treat it. Topics: Analgesics; Analysis of Variance; Animals; Anisoles; Disease Models, Animal; Dose-Response Relationship, Drug; gamma-Aminobutyric Acid; Hyperalgesia; Imidazolines; Male; Mice; Moclobemide; Monoamine Oxidase; Neuralgia; Pain Measurement; Pain Threshold; Pregabalin; Rotarod Performance Test; Time Factors | 2013 |
A back translation of pregabalin and carbamazepine against evoked and non-evoked endpoints in the rat spared nerve injury model of neuropathic pain.
The purpose of the present study was twofold. First to characterize endpoints distinct to the reflexive responses to sensory stimuli typically used in neuropathic pain models. A second aim was to evaluate two clinically approved drugs carbamazepine (Tegretol) and pregabalin (Lyrica) against these endpoints with the purpose to backtranslate from the clinical to preclinical setting. The selected neuropathic pain model was the spared nerve injury (SNI) model and the endpoints were burrowing and measures of paw posture in Sprague Dawley rats. As previously described, SNI surgery produced a robust heightened sensitivity to tactile and thermal (cold) stimuli. SNI surgery also produced robust decreases in burrowing and affected multiple measures of paw position. There was no correlation between magnitude of change in burrowing and sensory allodynia within SNI operated rats. Pregabalin (10-30 mg/kg IP) produced a reliable reversal of both tactile and cold allodynia and also the burrowing deficit, with minimal effect on neurological function evaluated using rotorod, beam walking and open field activity. Pregabalin did not affect any measure of paw position. Pharmacokinetic studies conducted in satellite animals identified plasma levels of pregabalin at the 10 mg/kg IP dose to be equivalent to clinically efficacious levels recorded in neuropathic patients (3-6 μg/ml). In contrast carbamazepine (10-60 mg/kg IP) had only a very modest effect against a reflexive (tactile) measure, and no effect against the burrowing deficit. Carbamazepine also affected various measures of neurological function, complicating interpretation of the reflexive measure. Measurement of burrowing appears to detect a behavioural deficit associated with the SNI model, that may be attenuated by pregabalin but not carbamazepine. Overall the present findings support an advantage of pregabalin over carbamazepine in terms of both efficacy and tolerability which is consistent with clinical experience. The inclusion of additional endpoints beyond traditional reflexive behaviours further supports the value of rodent neuropathic pain models, such as the SNI, as behavioural assays to detect new chemical entities to treat this pain condition. Topics: Animals; Behavior, Animal; Carbamazepine; Disease Models, Animal; Dose-Response Relationship, Drug; Gait; gamma-Aminobutyric Acid; Hyperalgesia; Male; Motor Activity; Neuralgia; Peripheral Nerve Injuries; Physical Stimulation; Postural Balance; Pregabalin; Rats; Rotarod Performance Test | 2013 |
Pregabalin attenuates excitotoxicity in diabetes.
Diabetes can exacerbate seizures and worsen seizure-related brain damage. In the present study, we aimed to determine whether the standard antiepileptic drug pregabalin (PGB) protects against pilocarpine-induced seizures and excitotoxicity in diabetes. Adult male Sprague-Dawley rats were divided into either a streptozotocin (STZ)-induced diabetes group or a normal saline (NS) group. Both groups were further divided into subgroups that were treated intravenously with either PGB (15 mg/kg) or a vehicle; all groups were treated with subcutaneous pilocarpine (60 mg/kg) to induce seizures. To evaluate spontaneous recurrent seizures (SRS), PGB-pretreated rats were fed rat chow containing oral PGB (450 mg) for 28 consecutive days; vehicle-pretreated rats were fed regular chow. SRS frequency was monitored for 2 weeks from post-status epilepticus day 15. We evaluated both acute neuronal loss and chronic mossy fiber sprouting in the CA3 area. In addition, we performed patch clamp recordings to study evoked excitatory postsynaptic currents (eEPSCs) in hippocampal CA1 neurons for both vehicle-treated rats with SRS. Finally, we used an RNA interference knockdown method for Kir6.2 in a hippocampal cell line to evaluate PGB's effects in the presence of high-dose ATP. We found that compared to vehicle-treated rats, PGB-treated rats showed less severe acute seizure activity, reduced acute neuronal loss, and chronic mossy fiber sprouting. In the vehicle-treated STZ rats, eEPSC amplitude was significantly lower after PGB administration, but glibenclamide reversed this effect. The RNA interference study confirmed that PGB could counteract the ATP-sensitive potassium channel (KATP)-closing effect of high-dose ATP. By opening KATP, PGB protects against neuronal excitotoxicity, and is therefore a potential antiepileptogenic in diabetes. These findings might help develop a clinical algorithm for treating patients with epilepsy and comorbid metabolic disorders. Topics: Adenosine Triphosphate; Animals; Anticonvulsants; Blood Glucose; Cell Line; Diabetic Neuropathies; Disease Models, Animal; Excitatory Postsynaptic Potentials; gamma-Aminobutyric Acid; Glyburide; Hippocampus; KATP Channels; Male; Neurons; Pilocarpine; Potassium Channels, Inwardly Rectifying; Pregabalin; Rats; Seizures; Streptozocin | 2013 |
Synergistic effects of celecoxib and bupropion in a model of chronic inflammation-related depression in mice.
This study was aimed to characterize the depression-like behaviour in the classical model of chronic inflammation induced by Complete Freund's Adjuvant (CFA). Male Swiss mice received an intraplantar (i.pl.) injection of CFA (50 µl/paw) or vehicle. Behavioural and inflammatory responses were measured at different time-points (1 to 4 weeks), and different pharmacological tools were tested. The brain levels of IL-1β and BDNF, or COX-2 expression were also determined. CFA elicited a time-dependent edema formation and mechanical allodynia, which was accompanied by a significant increase in the immobility time in the tail suspension (TST) or forced-swimming (FST) depression tests. Repeated administration of the antidepressants imipramine (10 mg/kg), fluoxetine (20 mg/kg) and bupropion (30 mg/kg) significantly reversed depression-like behaviour induced by CFA. Predictably, the anti-inflammatory drugs dexamethasone (0.5 mg/kg), indomethacin (10 mg/kg) and celecoxib (30 mg/kg) markedly reduced CFA-induced edema. The oral treatment with the analgesic drugs dipyrone (30 and 300 mg/kg) or pregabalin (30 mg/kg) significantly reversed the mechanical allodyinia induced by CFA. Otherwise, either dipyrone or pregabalin (both 30 mg/kg) did not significantly affect the paw edema or the depressive-like behaviour induced by CFA, whereas the oral treatment with dipyrone (300 mg/kg) was able to reduce the immobility time in TST. Noteworthy, CFA-induced edema was reduced by bupropion (30 mg/kg), and depression behaviour was prevented by celecoxib (30 mg/kg). The co-treatment with bupropion and celecoxib (3 mg/kg each) significantly inhibited both inflammation and depression elicited by CFA. The same combined treatment reduced the brain levels of IL-1β, as well as COX-2 immunopositivity, whilst it failed to affect the reduction of BDNF levels. We provide novel evidence on the relationship between chronic inflammation and depression, suggesting that combination of antidepressant and anti-inflammatory agents bupropion and celecoxib might represent an attractive therapeutic strategy for depression. Topics: Analgesics; Animals; Anti-Inflammatory Agents; Antidepressive Agents; Behavior, Animal; Brain-Derived Neurotrophic Factor; Bupropion; Celecoxib; Cerebral Cortex; Cyclooxygenase 2; Depression; Dipyrone; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Edema; Freund's Adjuvant; gamma-Aminobutyric Acid; Inflammation; Interleukin-1beta; Male; Mice; Nociception; Pregabalin; Pyrazoles; Sulfonamides | 2013 |
Antinociceptive effect of Butea monosperma on vincristine-induced neuropathic pain model in rats.
Neuropathic pain is a chronic neurodegenerative disease. It is well characterized by spontaneous pain, hyperalgesia, hypothesia, dysesthesia and allodynia. The present study was designed to investigate the antinociceptive potential of Butea monosperma on vincristine-induced painful neuropathy in rats. Vincristine was administered for induction of neuropathic pain in experimental animals. Hot plate, acetone drop, paw pressure, Von Frey hair and tail immersion tests were performed to assess the degree of thermal hyperalgesia, cold chemical allodynia, mechanical hyperalgesia and allodynia in the hind paw and tail thermal hyperalgesia, respectively, as an index of peripheral and central pain sensation. Tissue thiobarbituric acid reactive substances (TBARSs), reduced glutathione (GSH) and total calcium levels were estimated to assess the biochemical changes in the sciatic nerve tissue. Microscopically, histopathological changes were also observed in the sciatic nerve tissue. Ethanolic extract of B. monosperma leaves and pregabalin were administered for 14 consecutive days. Vincristine administration resulted in significant reduction in behavioural (i.e. hyperalgesia and allodynic pain sensation) changes along with a rise in the levels of TBARS, total calcium and decrease in GSH levels when compared with the normal control group. Moreover, significant histological changes were also observed. Pretreatment with B. monosperma significantly attenuated vincristine-induced development of painful behavioural, biochemical and histological changes in a dose-dependent manner, which is similar to that of pregabalin-pretreated group. B. monosperma ameliorated vincristine-induced painful neuropathy. It may be due to its potential of antioxidative, neuroprotective and calcium channel inactivation. Topics: Analgesics; Animals; Antineoplastic Agents, Phytogenic; Antioxidants; Behavior, Animal; Butea; Calcium; Disease Models, Animal; Female; gamma-Aminobutyric Acid; Glutathione; Hindlimb; Hot Temperature; Hyperalgesia; Male; Neuralgia; Oxidative Stress; Pain Measurement; Peripheral Nervous System Diseases; Plant Extracts; Plant Leaves; Pregabalin; Rats; Rats, Wistar; Sciatic Nerve; Tail; Thiobarbituric Acid Reactive Substances; Treatment Outcome; Vincristine | 2013 |
Intrathecal ketamine and pregabalin at sub-effective doses synergistically reduces neuropathic pain without motor dysfunction in mice.
Peripheral or central nerve injury often leads to neuropathic pain. Although ketamine and pregabalin are first line options for the treatment of neuropathic pain, their clinical application is limited due to side effects such as sedation, dizziness and somnolence. We designed this study to determine whether the intrathecal (i.t.) co-treatment with ketamine and pregabalin at sub-effective low doses would elicit a sufficient pain relief without producing side effect in a neuropathic pain mouse model. At day 7 after chronic constriction injury (CCI) of sciatic nerve, dose dependent effects of i.t. ketamine (3, 10, 30, 100 µg) or i.t. pregabalin (10, 30, 100 µg) on mechanical allodynia and thermal hyperalgesia were measured. For combination treatment, 3 or 10 µg of ketamine and 30 µg of pregabalin were selected because these doses of drugs were not effective on neuropathic pain. Interestingly, combined i.t. treatment groups (ketamine 3 µg+pregabalin 30 µg and ketamine 10 µg+pregabalin 30 µg) produced strong analgesia on neuropathic pain although these doses of ketamine and pregabalin alone are not effective. Moreover, rota rod test revealed that normal motor function was not affected by combined treatment while i.t. ketamine at doses above 10 µg showed a significant motor dysfunction. Results of this study suggested that i.t. co-treatment with ketamine and pregabalin at sub-effect low doses may be a useful therapeutic method for the treatment of neuropathic pain patients. Topics: Analgesics; Animals; Disease Models, Animal; Drug Synergism; gamma-Aminobutyric Acid; Hyperalgesia; Injections, Spinal; Ketamine; Male; Mice; Mice, Inbred ICR; Motor Skills; Neuralgia; Pregabalin | 2013 |
Combined antiallodynic effect of Neurotropin® and pregabalin in rats with L5-spinal nerve ligation.
In this study, we investigated the combined effect of Neurotropin® and pregabalin for L5-spinal nerve ligation (L5-SNL) model in rats and thiopental-induced sleep in mice.. The left fifth lumbar nerve of rats was tightly ligated with silk sutures under pentobarbital anesthesia. The hindpaw withdrawal threshold was measured by application of von Frey filaments. Thiopental sodium was intravenously administered in mice and sleeping time was measured. In L5-SNL rats, an isobolographic analysis was performed to clarify the combined antiallodynic effect of Neurotropin and pregabalin 14 days after ligation in rats. In isobolographic analysis and thiopental-induced sleep test, Neurotropin and pregabalin were orally administered to coincide with the timing of the peak effect of each drug.. Neurotropin (50-200 NU/kg) and pregabalin (2.5-10mg/kg) showed a dose-dependent antiallodynic action in L5-SNL rats. The antiallodynic effect of pregabalin was reversed by intrathecal injection of yohimbine or ondansetron. Isobolographic analysis suggested that the combined antiallodynic effect of Neurotropin and pregabalin in L5-SNL rats may have been more than a mere additive effect. Neurotropin (50-400 NU/kg) had no effect on thiopental-induced sleeping time whereas pregabalin (30-100mg/kg) significantly prolonged it. When the dose of pregabalin was 30 mg/kg, Neurotropin (50-400 NU/kg) did not further exacerbate the prolongation effect of pregabalin on thiopental-induced sleep.. It was suggested that when Neurotropin was administered in combination with pregabalin, it might provide more effective pain relief than that obtained with each agent alone in neuropathic pain without aggravating adverse effects of pregabalin. Topics: Administration, Oral; Analgesics; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Therapy, Combination; gamma-Aminobutyric Acid; Hyperalgesia; Ligation; Lumbosacral Region; Male; Neuralgia; Pain Threshold; Polysaccharides; Pregabalin; Rats; Rats, Wistar; Sleep; Spinal Nerves; Thiopental | 2013 |
Effects of pregabalin on behavioral alterations induced by ketamine in rats.
The aim of this study is to investigate the effects of pregabalin on the behavior of rats under the influence of ketamine, an NMDA receptor antagonist that mimics the symptoms of schizophrenia.. Rats were injected with saline or 25 mg/kg ketamine intraperitoneally. After that, behavior modifications were investigated by the evaluation of stereotypy and hyperlocomotion, after treating rats with pregabalin (at doses of 30 mg/kg or 100 mg/kg) or placebo (saline solution).. The administration of pregabalin reduced ketamine-induced hyperlocomotion. However, neither doses of pregabalin had a significant effect on ketamineinduced stereotypy.. This is the first study to investigate the effects of pregabalin using an animal model of psychosis. Furthermore, our results indicate that behavioral changes induced by ketamine in rats can be reversed with the use of pregabalin, suggesting its potential to treat psychotic symptoms. Topics: Animals; Antipsychotic Agents; Behavior, Animal; Disease Models, Animal; Dose-Response Relationship, Drug; Excitatory Amino Acid Antagonists; gamma-Aminobutyric Acid; Ketamine; Male; Pregabalin; Psychoses, Substance-Induced; Rats; Rats, Wistar; Schizophrenia | 2012 |
Interactions of pregabalin with gabapentin, levetiracetam, tiagabine and vigabatrin in the mouse maximal electroshock-induced seizure model: a type II isobolographic analysis.
The aim of this study was to characterize the anticonvulsant effects of pregabalin in combination with four second-generation antiepileptic drugs (i.e., gabapentin, levetiracetam, tiagabine, and vigabatrin) in the mouse maximal electroshock (MES)-induced seizure model by using the type II isobolographic analysis. Tonic hind limb extension (seizure activity) was evoked in adult male albino Swiss mice by a current (sine-wave, 25 mA, 500 V, 50 Hz, 0.2 s stimulus duration) delivered via auricular electrodes. The combination of pregabalin with gabapentin at the fixed-ratio of 1:1 was supra-additive (synergistic) in terms of seizure suppression while the combinations at the fixed-ratios of 2:1 and 4:1 were additive in the mouse MES model. Similarly, the combination of pregabalin with tiagabine at the fixed-ratio of 25:1 was supra-additive, whereas the combinations at the fixed-ratios of 100:1 and 50:1 were additive in the mouse MES model. Pregabalin with levetiracetam and vigabatrin at the fixed-ratios of 1:1, 2:1 and 4:1 were additive in this seizure model. The combinations of pregabalin with gabapentin (1:1) and pregabalin with tiagabine (25:1) appear to be favorable combinations exerting supra-additive interaction in suppressing MES-induced seizures. Pregabalin in combination with levetiracetam and vigabatrin appears to be neutral producing only additivity in the mouse MES model. Topics: Amines; Animals; Anticonvulsants; Avoidance Learning; Confidence Intervals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Electroshock; Gabapentin; gamma-Aminobutyric Acid; Hand Strength; Levetiracetam; Male; Mice; Motor Activity; Piracetam; Pregabalin; Seizures; Vigabatrin | 2012 |
Comparison of central versus peripheral delivery of pregabalin in neuropathic pain states.
Although pregabalin therapy is beneficial for neuropathic pain (NeP) by targeting the CaVα2δ-1 subunit, its site of action is uncertain. Direct targeting of the central nervous system may be beneficial for the avoidance of systemic side effects.. We used intranasal, intrathecal, and near-nerve chamber forms of delivery of varying concentrations of pregabalin or saline delivered over 14 days in rat models of experimental diabetic peripheral neuropathy and spinal nerve ligation. As well, radiolabelled pregabalin was administered to determine localization with different deliveries. We evaluated tactile allodynia and thermal hyperalgesia at multiple time points, and then analyzed harvested nervous system tissues for molecular and immunohistochemical changes in CaVα2δ-1 protein expression. Both intrathecal and intranasal pregabalin administration at high concentrations relieved NeP behaviors, while near-nerve pregabalin delivery had no effect. NeP was associated with upregulation of CACNA2D1 mRNA and CaVα2δ-1 protein within peripheral nerve, dorsal root ganglia (DRG), and dorsal spinal cord, but not brain. Pregabalin's effect was limited to suppression of CaVα2δ-1 protein (but not CACNA2D1 mRNA) expression at the spinal dorsal horn in neuropathic pain states. Dorsal root ligation prevented CaVα2δ-1 protein trafficking anterograde from the dorsal root ganglia to the dorsal horn after neuropathic pain initiation.. Either intranasal or intrathecal pregabalin relieves neuropathic pain behaviours, perhaps due to pregabalin's effect upon anterograde CaVα2δ-1 protein trafficking from the DRG to the dorsal horn. Intranasal delivery of agents such as pregabalin may be an attractive alternative to systemic therapy for management of neuropathic pain states. Topics: Animals; Behavior, Animal; Blotting, Western; Calcium Channels; Calcium Channels, L-Type; Central Nervous System; Diabetic Neuropathies; Disease Models, Animal; Drug Administration Routes; gamma-Aminobutyric Acid; Ganglia, Spinal; Gene Expression Regulation; Immunohistochemistry; Ligation; Male; Microglia; Neuralgia; Peripheral Nervous System; Pregabalin; Rats; Rats, Sprague-Dawley; Reproducibility of Results; RNA, Messenger; Spinal Nerve Roots; Spinal Nerves | 2012 |
Pharmacological characterization of lysophosphatidic acid-induced pain with clinically relevant neuropathic pain drugs.
Lysophosphatidic acid (LPA), an initiator of neuropathic pain, causes allodynia. However, few studies have evaluated the pharmacological profile of LPA-induced pain. In this study, a LPA-induced pain model was developed and pharmacologically characterized with clinically relevant drugs used for neuropathic pain, including antiepileptics, non-steroidal anti-inflammatory agents, analgesics, local anaesthetics/antiarrhythmics and antidepressants. Gabapentin (1-30 mg/kg, p.o.) significantly reversed LPA-induced allodynia, but neither indomethacin (30 mg/kg, p.o.) nor morphine (0.3-3 mg/kg, s.c.) did, which indicates that LPA-induced pain consists mostly of neuropathic rather than inflammatory pain. Both pregabalin (0.3-10 mg/kg, p.o.) and ω-CgTX MVIIA (0.01-0.03 μg/mouse, i.t.) completely reversed LPA-induced allodynia in a dose-dependent manner. Lidocaine (1-30 mg/kg, s.c.), mexiletine (1-30 mg/kg, p.o.) and carbamazepine (10-100 mg/kg, p.o.) significantly ameliorated LPA-induced allodynia dose dependently. Milnacipran (30 mg/kg, i.p.) produced no significant analgesic effect in LPA-induced allodynia. In LPA-injected mice, expression of the α2δ1 subunit of the voltage-gated calcium channel (VGCC) was increased in the dorsal root ganglion (DRG) and spinal dorsal horn. Furthermore, the VGCC current was potentiated in both the DRG from LPA-injected mice and LPA (1 μM)-treated DRG from saline-injected mice, and the potentiated VGCC current was amended by treatment with gabapentin (100 μM). The LPA-induced pain model described here mimics aspects of the neuropathic pain state, including the sensitization of VGCC, and may be useful for the early assessment of drug candidates to treat neuropathic pain. Topics: Amines; Analgesics; Animals; Calcium Channels; Cyclohexanecarboxylic Acids; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Ganglia, Spinal; Hyperalgesia; Indomethacin; Lysophospholipids; Male; Mice; Morphine; Motor Activity; Neuralgia; Neurons; Pregabalin; Spinal Cord | 2012 |
The efficacy of morphine, pregabalin, gabapentin, and duloxetine on mechanical allodynia is different from that on neuroma pain in the rat neuropathic pain model.
It has been reported that <50% of neuropathic pain patients are satisfactorily treated with drugs. It is possible that this lack of efficacy of drugs on neuropathic pain might be due to the drugs prescribed, regardless of the origin of pain. We compared the efficacy of orally administered morphine, pregabalin, gabapentin, and duloxetine on mechanical allodynia with that on neuroma pain using the tibial neuroma transposition (TNT) model.. In the TNT model, the tibial nerve is transected, and the tibial nerve stump is transpositioned to the lateral aspect of the hindlimb. After TNT injury, mechanical allodynia and neuroma pain are observed. Morphine, pregabalin, gabapentin, and duloxetine were administered orally and were examined for the antiallodynic and antineuroma pain effects.. Morphine, pregabalin, gabapentin, and duloxetine attenuated the level of mechanical allodynia in a dose-dependent manner. Morphine-but not pregabalin, gabapentin, and duloxetine-attenuated the neuroma pain. Morphine was less potent in neuroma pain than in mechanical allodynia. In the 2-drug-combination studies (morphine + pregabalin, morphine + duloxetine, and pregabalin + duloxetine), all drug combinations produced a synergistic effect on mechanical allodynia, but not on neuroma pain.. These data indicate that the potency of morphine and the efficacy of pregabalin, gabapentin, and duloxetine on mechanical allodynia are different from those on neuroma pain and that combination therapy is one of different therapeutic choices for the treatment of neuropathic pain. Topics: Administration, Oral; Amines; Analgesics, Non-Narcotic; Analgesics, Opioid; Animals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Drug Therapy, Combination; Duloxetine Hydrochloride; Gabapentin; gamma-Aminobutyric Acid; Hyperalgesia; Male; Morphine; Neuralgia; Neuroma; Pain Measurement; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Thiophenes; Time Factors | 2012 |
Etodolac, a cyclooxygenase-2 inhibitor, attenuates paclitaxel-induced peripheral neuropathy in a mouse model of mechanical allodynia.
The effect of the cyclooxygenase-2 (COX-2) inhibitor etodolac on the mechanical allodynia induced by paclitaxel was investigated in mice and compared with the effects of the nonselective COX inhibitors indomethacin and diclofenac, the selective COX-2 inhibitor celecoxib, the calcium channel α(2)δ subunit inhibitor pregabalin, the sodium channel blocker mexiletine, and the serotonin-norepinephrine reuptake inhibitor duloxetine. The decrease in the paw-withdrawal threshold induced by paclitaxel was reversed by oral administration of etodolac at 10 mg/kg but was not affected by indomethacin, diclofenac, or celecoxib. The antiallodynic effect of etodolac gradually increased during repeated administration, and after 2 weeks the paw-withdrawal threshold at the preadministration point was significantly increased. Pregabalin, duloxetine, and mexiletine also showed an antiallodynic effect in this model. Whereas pregabalin had a preadministration effect similar to that of etodolac during repeated administration, mexiletine or duloxetine had no such effect. There was almost no difference in the distribution of etodolac and diclofenac in nervous tissue, indicating that COX inhibition is unlikely to be involved in the antiallodynic effect of etodolac. Etodolac did not show a neuroprotective effect against morphological transformations such as the axonal degeneration induced by paclitaxel. Instead, etodolac probably acts at the level of functional changes accompanying paclitaxel treatment, such as alterations in the activation state of components of the pain transmission pathway. Our findings suggest that etodolac attenuates paclitaxel-induced peripheral neuropathy by a COX-independent pathway and that it might be useful for the treatment of paclitaxel-induced peripheral neuropathy. Topics: Animals; Cyclooxygenase 2; Cyclooxygenase 2 Inhibitors; Diclofenac; Disease Models, Animal; Drug Interactions; Duloxetine Hydrochloride; Etodolac; gamma-Aminobutyric Acid; Hyperalgesia; Male; Mexiletine; Mice; Paclitaxel; Peripheral Nervous System Diseases; Pregabalin; Thiophenes; Tissue Distribution | 2012 |
Effect of the anticonvulsant medications pregabalin and lamotrigine on urodynamic parameters in an animal model of neurogenic detrusor overactivity.
To assess the effects of different doses and treatment durations of pregabalin and lamotrigine on the urodynamic parameters of an animal model of neurogenic detrusor overactivity (NDO).. Ninety rats were used; six as normal controls and the remaining 84 were divided as follows: Six "paraplegic controls," 6 "paraplegic-vehicle controls," and the remaining 72 divided into two equal groups. Group 1 was divided into six subgroups; pregabalin was given in doses of 10 mg/kg, 20 mg/kg, or 30 mg/kg for 1 or 2 weeks. Group 2 was similarly subdivided; lamotrigine was given in doses of 1.5 mg/kg, 3 mg/kg, or 6 mg/kg for 1 or 2 weeks.. All paraplegic controls developed NDO within 3 weeks from spinalization. Their baseline bladder pressure (BBP) 19 ± 4.4 cmH(2) O, detrusor pressure at maximum capacity (DPMaxC) 47.6 ± 4.3 cmH(2) O, bladder capacity (BC) 0.45 ± 0.1 ml, and frequency of detrusor overactivity (FDO) 3.7 ± 0.9/min. Both pregabalin and lamotrigine produced significant improvement. Urodynamic values in those treated with 20 mg pregabalin for 1 or 2 weeks were: BBP 11.7 ± 1.3 and 9 ± 0.2 cmH(2) O, BC 0.6 ± 0.1 and 0.7 ± 0.01 ml, DPMaxC 17.3 ± 4.0 and 23 ± 2.6 cmH(2) O, FDO 2.1 ± 0.2/min and 1.7 ± 0.1/min. Urodynamic values in those treated with 3 mg/kg lamotrigine for 1 or 2 weeks were: BBP 9.7 ± 2.2 and 8.6 ± 1.9 cmH(2) O, DPMaxC 17.2 ± 1.8 and 29 ± 1.2 cmH(2) O, BC 0.7 ± 0.1 and 0.8 ± 0.1 ml, FDO 1.9 ± 0.2/min and 1.9 ± 0.2/min (P < 0.001).. Pregabalin and lamotrigine may represent novel alternative treatments of NDO. Clinical trials remain to be performed. Topics: Animals; Anticonvulsants; Disease Models, Animal; Dose-Response Relationship, Drug; Female; gamma-Aminobutyric Acid; Lamotrigine; Paraplegia; Pregabalin; Pressure; Rats; Rats, Sprague-Dawley; Time Factors; Triazines; Urinary Bladder; Urinary Bladder, Neurogenic; Urinary Bladder, Overactive; Urination; Urodynamics | 2012 |
Systemic pregabalin attenuates sensorimotor responses and medullary glutamate release in inflammatory tooth pain model.
Our previous studies have demonstrated that application of inflammatory irritant mustard oil (MO) to the tooth pulp induces medullary glutamate release and central sensitization in the rat medullary dorsal horn (MDH), as well as nociceptive sensorimotor responses in craniofacial muscles in rats. There is recent evidence that anticonvulsant drugs such as pregabalin that influence glutamatergic neurotransmission are effective in several pain states. The aim of this study was to examine whether systemic administration of pregabalin attenuated glutamate release in the medulla as well as these nociceptive effects reflected in increased electromyographic (EMG) activity induced by MO application to the tooth pulp. Male adult rats were anesthetized with isofluorane (1.0-1.2%), and jaw and tongue muscle EMG activities were recorded by needle electrodes inserted bilaterally into masseter and anterior digastric muscles and into the genioglossus muscle, and also the medullary release of glutamate was assessed by in vivo microdialysis. Pregabalin or vehicle control (isotonic saline) was administered 30 min before the pulpal application of MO or vehicle control (mineral oil). Application of mineral oil to the maxillary first molar tooth pulp produced no change in baseline EMG activity and glutamate release. However, application of MO to the pulp significantly increased both the medullary release of glutamate and EMG activity in the jaw and tongue muscles for several minutes. In contrast, pre-medication with pregabalin, but not vehicle control, significantly and dose-dependently attenuated the medullary glutamate release and EMG activity in these muscles after MO application to the tooth pulp (analysis of variance (ANOVA), p<0.05). These results suggest that pregabalin may attenuate the medullary release of glutamate and associated nociceptive sensorimotor responses in this acute inflammatory pulpal pain model, and that it may prove useful for the treatment of orofacial inflammatory pain states. Topics: Analgesics; Animals; Dental Pulp; Disease Models, Animal; Electromyography; Facial Muscles; gamma-Aminobutyric Acid; Glutamic Acid; Inflammation; Irritants; Male; Medulla Oblongata; Microdialysis; Mustard Plant; Plant Oils; Pregabalin; Rats; Rats, Sprague-Dawley; Toothache | 2012 |
Pregabalin antinociception and its interaction with tramadol in acute model of pain.
The aim of present study was to investigate the antinociceptive effect of pregabalin and tramadol either alone and or in combination on acute model of pain.. The antinociceptive effect of intraperitoneal administration of pregabalin (1 to 400 mg/kg) and tramadol (10 to 80 mg/kg) or combination of them were measured after 30 and 60 min on hot-plate in terms of maximum possible effect (%MPE) in mice.. Antinociceptive effect rose significantly for both pregabalin at doses 200 and 400 mg/kg and tramadol from 20 to 80 mg/kg in dose dependent manner. From linear equation the doses that increased antinociceptive effect by 50% (ED(50)) were 69 ± 8.2 mg/kg for tramadol and 246 ± 24 mg/kg for pregabalin. Unlike pregabalin, %MPE(30) (at 30(th) min) of tramadol was significantly higher than its %MPE(60). The interaction after co-administration of non analgesic dose of 10 mg/kg of pregabalin with low analgesic dose of 30 mg/kg of tramadol resulted super-additive and %MPE(30) and %MPE(60) were increased compared to each drug alone. In all other combination groups, the interaction were sub-additive particularly when non analgesic doses of each drug (10 mg/kg) were co-administrated and %MPE was decreased significantly compared to that of each drug alone.. Pregabalin revealed a comparative antinociceptive effect as similar to tramadol in acute model of pain, but interaction between these two drugs depends highly on their proportion in the combination. The analgesia may increase but adverse effects such as seizurogenic effect of tramadol can be reduced in clinical setting if right proportion is used. More studies are required to understand the mechanisms and clinical implication of such combinations. Topics: Acute Pain; Analgesics; Analgesics, Opioid; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Drug Synergism; Drug Therapy, Combination; gamma-Aminobutyric Acid; Injections, Intraperitoneal; Male; Mice; Pain; Pregabalin; Time Factors; Tramadol | 2012 |
Synergistic interaction of pregabalin with the synthetic cannabinoid WIN 55,212-2 mesylate in the hot-plate test in mice: an isobolographic analysis.
The aim of the study was to determine the type of interaction between pregabalin (a 3(rd)-generation antiepileptic drug) and WIN 55,212-2 mesylate (WIN - a highly potent non-selective cannabinoid CB1 and CB2 receptor agonist) administered in combination at a fixed ratio of 1:1, in the acute thermal pain model (hot-plate test) in mice.. Linear regression analysis was used to evaluate the dose-response relationships between logarithms of drug doses and their resultant maximum possible antinociceptive effects in the mouse hot-plate test. From linear equations, doses were calculated that increased the antinociceptive effect by 30% (ED(30) values) for pregabalin, WIN, and their combination. The type of interaction between pregabalin and WIN was assessed using the isobolographic analysis.. Results indicated that both compounds produced a definite antinociceptive effect, and the experimentally-derived ED(30) values for pregabalin and WIN, when applied alone, were 29.4 mg/kg and 10.5 mg/kg, respectively. With isobolography, the experimentally derived ED(30 mix) value for the fixed ratio combination of 1:1 was 5.7 mg/kg, and differed significantly from the theoretically calculated ED(30 add) value of 19.95 mg/kg (p < 0.01), indicating synergistic interaction between pregabalin and WIN in the hot-plate test in mice.. Isobolographic analysis demonstrated that the combination of WIN with pregabalin at a fixed ratio of 1:1 exerted synergistic interaction in the mouse model of acute thermal pain. If the results from this study could be adapted to clinical settings, the combination of WIN with pregabalin might be beneficial for pain relief in humans. Topics: Acute Pain; Analgesics; Animals; Benzoxazines; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; gamma-Aminobutyric Acid; Linear Models; Male; Mice; Morpholines; Naphthalenes; Pregabalin; Receptor, Cannabinoid, CB1; Receptor, Cannabinoid, CB2 | 2012 |
Combined carbamazepine and pregabalin therapy in a rat model of neuropathic pain.
Carbamazepine and pregabalin have proven effects against neuropathic pain. Carbamazepine blocks voltage-dependent Na(+) channels, whereas pregabalin blocks voltage-dependent Ca(2+) channels. The authors hypothesized that the co-administration of these drugs would synergistically reduce neuropathic pain.. Neuropathic pain was induced by L5 nerve ligation in Sprague-Dawley rats. To determine their ED(50) values, carbamazepine and pregabalin were orally administered at 0.3, 3, 10, or 30 mg kg(-1). The drugs were then co-administered at 0, 1/4×ED(50), 1/2×ED(50), 1.5×ED(50), and 2×ED(50) to determine the ED(50) and ED(75) values of the drugs in combination. Allodynia was determined using the von Frey hair test and dose-effect curves and isobolograms were used to investigate drug interactions. Levels of the acute reactive protein c-Fos in the dorsal horn were evaluated as an indicator of pathological nerve excitation.. At ED(50) levels, carbamazepine and pregabalin did not exhibit synergism, but doses higher than ED(75) were found to be synergistic. The combination index was 0.18 (strong synergy) and dose reductions were 35.7-fold for carbamazepine and 6.8-fold for pregabalin when co-administered when compared with a single administration at ED(75). The percentage allodynia relief was only 60% for carbamazepine and 80% for pregabalin by single administration, whereas their co-administration relieved allodynia by 100%. Furthermore, treatment decreased c-Fos expression in the dorsal horn, but expressional differences between animals treated with carbamazepine plus pregabalin were not significantly different from those treated with single drug.. Carbamazepine and pregabalin ameliorate neuropathic pain synergistically at higher doses. Topics: Analgesics; Animals; Carbamazepine; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Drug Therapy, Combination; gamma-Aminobutyric Acid; Hyperalgesia; Male; Neuralgia; Pain Measurement; Pregabalin; Rats; Rats, Sprague-Dawley | 2012 |
Pregabalin attenuates place escape/avoidance behavior in a rat model of spinal cord injury.
Spinal cord injury (SCI) pain in humans is difficult to treat, and the lack of valid methods to measure behavior comparable to the complex human pain experience preclinically represents an important obstacle to finding better treatments for this type of central pain. The place escape/avoidance paradigm (PEAP) relies on the active choice of an animal between its natural preference for a dark environment or pain relief, and it has been suggested to measure the affective-motivational component of pain. We have modified the method to a T10 spinal cord contusion model (SCC) of at-level central neuropathic pain in Sprague-Dawley rats. In order to demonstrate sensitivity to change in escape/avoidance behavior and thus the applicability of the PEAP method to predict drug efficacy, we investigated the effect of pregabalin (30 mg/kg) treatment in a randomized design. SCC animals displayed increased escape/avoidance behavior postinjury, indicating at-level mechanical hypersensitivity. Second, we found no correlation between state anxiety levels in SCC animals (elevated plus maze) and PEAP behavior, suggesting that the PEAP measurement is not biased by differences in anxiety levels. Third, we demonstrated a decrease in escape/avoidance behavior in response to treatment with the analgesic drug pregabalin. Thus, the PEAP may be applicable as a surrogate correlate of human pain. In conclusion, the primary finding in this study was a sensitivity to change in escape/avoidance behavior induced by pharmacological modulation with analgesics, supporting the use of the PEAP as a central outcome measure in preclinical SCI pain research. Topics: Analgesics; Animals; Anxiety; Avoidance Learning; Behavior, Animal; Disease Models, Animal; Fear; Female; gamma-Aminobutyric Acid; Neuropsychological Tests; Outcome Assessment, Health Care; Pain, Intractable; Pregabalin; Rats; Rats, Sprague-Dawley; Spinal Cord Injuries | 2011 |
Gabapentin and pregabalin inhibit the itch-associated response induced by the repeated application of oxazolone in mice.
We investigated the effects of gabapentin and pregabalin on the itch-associated response in a mouse model of chronic dermatitis induced by the repeated application of 4-ethoxymethylene-2-phenyl-2-oxazolin-5-one (oxazolone). Challenging the mice with oxazolone-induced chronic dermatitis with the oxazolone evoked severe and transient scratching behavior until 1 h after the application of oxazolone. Thereafter, a more mild and continuous scratching behavior was also observed for at least 8 h. Both severe and continuous scratching behaviors were suppressed by systemic injection of gabapentin and pregabalin. This effect of these compounds was correlated with its affinity for the α₂δ subunit of voltage-gated Ca²(+) channels. Intrathecal injection, but not peripheral treatment, with gabapentin inhibited the scratching behavior in this model. Gabapentin failed to suppress the scratching behavior induced by the intradermal injection of compound 48/80 in normal mice. The expression of the α₂δ-1 subunit in dorsal root ganglion (DRG) from mice following repeated application of oxazolone was significantly higher than that from normal mice. These results suggest that gabapentin and pregabalin show an anti-pruritic activity through α₂δ-subunit binding, and the up-regulation of the α₂δ-1 subunit in DRG may therefore play an important role in its anti-pruritic activity. Topics: Amines; Animals; Calcium Channels; Chronic Disease; Cyclohexanecarboxylic Acids; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Eruptions; Gabapentin; gamma-Aminobutyric Acid; Ganglia, Spinal; Injections, Spinal; Male; Mice; Mice, Inbred BALB C; Oxazolone; Pregabalin; Rats; Rats, Sprague-Dawley; Severity of Illness Index; Time Factors; Up-Regulation; Urticaria | 2011 |
7-Nitroindazole, but not NG-nitro-L-arginine, enhances the anticonvulsant activity of pregabalin in the mouse maximal electroshock-induced seizure model.
The objective of this study was to determine the effects of 7-nitroindazole (7NI--a preferential neuronal nitric oxide synthase (NOS) inhibitor) and NG-nitro-L-arginine (NNA--a non-selective NOS inhibitor) on the anticonvulsant action of pregabalin (PGB--a third-generation antiepileptic drug) in the maximal electroshock (MES)-induced seizure model in mice. Electroconvulsions were produced in mice by means of an alternating current (50 Hz, 500 V, 25 mA, ear-clip electrodes, 0.2 s stimulus duration, tonic hindlimb extension taken as the endpoint). The anticonvulsant action of PGB in the MES test was expressed as median effective doses (ED50 values) of the drug, protecting 50% of animals tested against MES-induced seizures. The acute adverse-effect potentials of PGB in combination with 7NI and NNA were evaluated in the chimney test (motor coordination), step-through passive avoidance task (long-term memory) and grip-strength test (skeletal muscular strength) in mice. 7NI (50 mg/kg, ip) significantly enhanced the anticonvulsant action of PGB by reducing the ED50 value of PGB from 145.0 mg/kg to 74.4 mg/kg (p<0.01). Similarly, 7NI at the lower dose of 25 mg/kg also potentiated the anticonvulsant action of PGB by lowering the ED50 value of PGB from 145.0 mg/kg to 117.9 mg/kg, although the results did not attain statistical significance. In contrast, NNA (40 mg/kg, ip) had no impact on the anticonvulsant effects of PGB. Moreover, none of the examined combinations of PGB with 7NI and NNA affected motor coordination, long-term memory and skeletal muscular strength in mice. Based on this preclinical study, one can conclude that 7NI significantly enhanced and NNA had no effect on the anticonvulsant activity of PGB against MES-induced seizures in mice. Topics: Animals; Anticonvulsants; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Drug Therapy, Combination; Electroshock; Enzyme Inhibitors; gamma-Aminobutyric Acid; Indazoles; Male; Mice; Neuroprotective Agents; Nitroarginine; Pregabalin; Seizures; Toxicity Tests, Acute | 2011 |
Synergistic antihypersensitive effects of pregabalin and tapentadol in a rat model of neuropathic pain.
Neuropathic pain is a clinical condition which remains poorly treated and combinations of pregabalin, an antagonist of the α2δ-subunit of Ca(2+) channels, with tapentadol, a μ-opioid receptor agonist/noradrenaline reuptake inhibitor, or with classical opioids such as oxycodone and morphine might offer increased therapeutic potential. In the rat spinal nerve ligation model, a dose dependent increase in ipsilateral paw withdrawal thresholds was obtained using an electronic von Frey filament after IV administration of pregabalin (1-10mg/kg), tapentadol (0.316-10mg/kg), morphine (1-4.64 mg/kg) and oxycodone (0.316-3.16 mg/kg), with ED(50) values (maximal efficacy) of 4.21 (67%), 1.65 (94%), 1.70 (96%) and 0.63 mg/kg (100%), respectively. Equianalgesic dose combinations of pregabalin and tapentadol (dose ratio 2.5:1), morphine (2.5:1) or oxycodone (6.5:1) resulted in ED(50) values (maximal efficacy) of 0.83 (89%), 2.33 (97%) and 1.14 mg/kg (100%), respectively. The concept of dose-equivalence suggested an additive interaction of pregabalin and either oxycodone or morphine, while a synergistic interaction was obtained with pregabalin and tapentadol (demonstrated by isobolographic analysis). There was no increase in contralateral paw withdrawal thresholds and no locomotor impairment, as measured in the open field, for the combination of pregabalin and tapentadol; while a significant increase and impairment was demonstrated for the combinations of pregabalin and either morphine or oxycodone. Because combination of pregabalin and tapentadol resulted in a synergistic antihypersensitive activity, it is suggested that, beside the use of either drug alone, this drug combination may offer a beneficial treatment option for neuropathic pain. Topics: Animals; Antihypertensive Agents; Behavior, Animal; Disease Models, Animal; Drug Synergism; gamma-Aminobutyric Acid; Ligation; Locomotion; Male; Morphine; Neuralgia; Oxycodone; Pain Threshold; Phenols; Pregabalin; Rats; Rats, Sprague-Dawley; Spinal Nerves; Tapentadol | 2011 |
Pregabalin modulation of spinal and brainstem visceral nociceptive processing.
Brainstem and spinal mechanisms mediating visceral nociception are investigated here using electrophysiology and immunohistochemistry techniques in a model of acute visceral pain. Colorectal distension (CRD) produced graded visceromotor responses (VMR) in normal rats, and these were facilitated by intracolonic mustard oil (MO) that generated acute visceral hyperalgesia. The neuropathic pain drug pregabalin (PGB) is thought to have state-dependent effects in attenuating neuropathic, but not acute somatic pain, likely by impairing calcium-channel trafficking. We found that systemic PGB produced antinociceptive effects on CRD-evoked VMRs in naïve rats lacking pathophysiology and in MO-pretreated rats. Systemic PGB also significantly reduced Fos labelling in lumbosacral spinal cords of rats given noxious repetitive CRD; however, PGB did not alter this measure of neural activity in the brainstem. Differential brainstem processing of noxious somatic and visceral stimuli may underlie the unique lack of state-dependent actions of PGB in this visceral pain model. Single-unit recordings in the rostral ventromedial medulla (RVM) verify that brainstem processing of somatic and visceral stimuli differs. The effects of CRD on RVM cells classed as ON, OFF, or NEUTRAL were independent of their somatic responses, with surprising changes in RVM cell activity to innocuous visceral stimulation. PGB also markedly reduced the visceral responses of RVM ON-cells to noxious CRD. These results illustrate clear differences in the central processing of visceral and somatic stimuli, yet a common role for descending modulation by brainstem activity in mediating evoked pain measures. Topics: Afferent Pathways; Analgesics; Animals; Brain Stem; Disease Models, Animal; gamma-Aminobutyric Acid; Male; Neuralgia; Nociceptors; Pregabalin; Rats; Rats, Sprague-Dawley; Spinal Cord; Visceral Pain | 2011 |
Characterization of a new animal model for evaluation and treatment of back pain due to lumbar facet joint osteoarthritis.
Osteoarthritic (OA) degeneration of the lumbar facet joints has been implicated in low back pain. This study was undertaken to investigate the biologic links between cellular and structural alterations within facet joint components and the development of symptomatic chronic back pain.. We generated an animal model of facet joint degeneration by intraarticular injection of monosodium iodoacetate (MIA) into facet joints (L3-L4, L4-L5, L5-L6) of Sprague-Dawley rats. Pain sensation due to pressure, which mimics a mechanical stimulus for facet joint injury, was measured using an algometer. Pain response was also assessed in a straight leg raising test. Cartilage alterations were assessed by biochemical evaluation and microfocal computed tomography (micro-CT). Therapeutic modulation of chronic facet joint pain with the use of various pharmacologic agents was investigated.. MIA injection resulted in severely damaged facet joint cartilage, proteoglycan loss, and alterations of subchondral bone structure. Micro-CT analyses suggested that the behavioral hyperalgesia from facet joint degeneration was not associated with foraminal stenosis. The biologic and structural changes in facet joints were closely associated with sustained and robust chronic pain. Morphine and pregabalin markedly alleviated pressure hyperalgesia, while celecoxib (a selective inhibitor of cyclooxygenase 2 [COX-2]) produced moderate antihyperalgesic effects and the effect of ketorolac (an inhibitor of COX-1 and COX-2) was negligible.. Our findings demonstrate that MIA injection provides a useful model for the study of OA changes in the facet joint and indicate that facet joint degeneration is a major cause of chronic low back pain. The treatment results suggest that classes of drugs that are widely used to treat OA, such as nonsteroidal antiinflammatory drugs, may have limited efficacy once joint destruction is complete. Topics: Analgesics; Animals; Back Pain; Cartilage, Articular; Disease Models, Animal; gamma-Aminobutyric Acid; Hyperalgesia; Morphine; Osteoarthritis, Spine; Pain Measurement; Pregabalin; Rats; Rats, Sprague-Dawley; Zygapophyseal Joint | 2011 |
The rat intervertebral disk degeneration pain model: relationships between biological and structural alterations and pain.
Degeneration of the interverterbral disk is as a cause of low-back pain is increasing. To gain insight into relationships between biological processes, structural alterations and behavioral pain, we created an animal model in rats.. Disk degeneration was induced by removal of the nucleus pulposus (NP) from the lumbar disks (L4/L5 and L5/L6) of Sprague Dawley rats using a 0.5-mm-diameter microsurgical drill. The degree of primary hyperalgesia was assessed by using an algometer to measure pain upon external pressure on injured lumbar disks. Biochemical and histological assessments and radiographs of injured disks were used for evaluation. We investigated therapeutic modulation of chronic pain by administering pharmaceutical drugs in this animal model.. After removal of the NP, pressure hyperalgesia developed over the lower back. Nine weeks after surgery we observed damaged or degenerated disks with proteoglycan loss and narrowing of disk height. These biological and structural changes in disks were closely related to the sustained pain hyperalgesia. A high dose of morphine (6.7 mg/kg) resulted in effective pain relief. However, high doses of pregabalin (20 mg/kg), a drug that has been used for treatment of chronic neuropathic pain, as well as the anti-inflammatory drugs celecoxib (50 mg/kg; a selective inhibitor of cyclooxygenase 2 (COX-2)) and ketorolac (20 mg/kg; an inhibitor of COX-1 and COX-2), did not have significant antihyperalgesic effects in our disk injury animal model.. Although similarities in gene expression profiles suggest potential overlap in chronic pain pathways linked to disk injury or neuropathy, drug-testing results suggest that pain pathways linked to these two chronic pain conditions are mechanistically distinct. Our findings provide a foundation for future research on new therapeutic interventions that can lead to improvements in the treatment of patients with back pain due to disk degeneration. Topics: Animals; Disease Models, Animal; gamma-Aminobutyric Acid; Intervertebral Disc Degeneration; Low Back Pain; Lumbar Vertebrae; Pain Measurement; Pregabalin; Rats; Rats, Sprague-Dawley | 2011 |
Synthesis and in vivo evaluation of 3,4-disubstituted gababutins.
A range of 3,4-alkylated five-membered ring derivatives of gabapentin were synthesised. One compound (21) had an excellent level of potency against alpha(2)delta and was profiled in in vivo models of pain and anxiety. Topics: Amines; Amino Acids; Animals; Anti-Anxiety Agents; Cyclohexanecarboxylic Acids; Cyclopentanes; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Hyperalgesia; Rats | 2010 |
Interactions of 1-methyl-1,2,3,4-tetrahydroisoquinoline with lamotrigine, oxcarbazepine, pregabalin, and topiramate in the mouse maximal electroshock-induced seizure model: a type I isobolographic analysis.
The aim of this study was to characterize the anticonvulsant effects of 1-methyl-1,2,3,4-tetrahydroisoquinoline (MeTHIQ--an endogenous parkinsonism-preventing substance) in combination with four second-generation antiepileptic drugs (AEDs: lamotrigine [LTG], oxcarbazepine [OXC], pregabalin [PGB], and topiramate [TPM]) in the mouse maximal electroshock (MES)-induced seizure model by using the type I isobolographic analysis for parallel and non-parallel dose-response relationship curves (DRRCs). Potential adverse-effect profiles of interactions of MeTHIQ with LTG, OXC, PGB and TPM at the fixed-ratio of 1:1 from the MES test with respect to motor performance, long-term memory and skeletal muscular strength were measured along with total brain concentrations of MeTHIQ and TPM. In the mouse MES model, MeTHIQ administered singly had its DRRC parallel to those for OXC and TPM, and simultaneously, non-parallel to those for LTG and PGB. With type I isobolography for parallel DRRCs, the combination of MeTHIQ with TPM at three fixed-ratios of 1:3, 1:1 and 3:1 exerted supra-additive (synergistic) interaction, whereas the combination of MeTHIQ with OXC at the fixed-ratios of 1:3, 1:1 and 3:1 produced additive interaction. Similarly, the type I isobolography for non-parallel DRRCs revealed that the combination of MeTHIQ with LTG and PGB at the fixed-ratio of 1:1 produced additive interaction. For all combinations, neither motor coordination, long-term memory nor muscular strength were affected. Total brain concentrations of MeTHIQ and TPM revealed no significant changes in their concentrations when the drugs were combined at the fixed-ratios of 1:3, 1:1 and 3:1. In conclusion, the synergistic interaction of MeTHIQ with TPM at the fixed-ratios of 1:3, 1:1 and 3:1 against MES-induced seizures was pharmacodynamic in nature and thus, it is worthy of consideration in further clinical settings. The combinations of MeTHIQ with LTG, OXC and PGB were neutral in the mouse MES model. Topics: Animals; Anticonvulsants; Avoidance Learning; Carbamazepine; Disease Models, Animal; Dopamine Antagonists; Dose-Response Relationship, Drug; Drug Administration Schedule; Drug Interactions; Drug Synergism; Drug Therapy, Combination; Electroshock; Fructose; gamma-Aminobutyric Acid; Lamotrigine; Male; Memory; Mice; Oxcarbazepine; Pregabalin; Seizures; Tetrahydroisoquinolines; Topiramate; Triazines | 2010 |
Evidence that pregabalin reduces neuropathic pain by inhibiting the spinal release of glutamate.
Pregabalin is an anti-convulsant that successfully treats many neuropathic pain syndromes, although the mechanism of its anti-hyperalgesic action remains elusive. This study aims to help delineate pregabalin's anti-hyperalgesic mechanisms. We assessed the effectiveness of pregabalin at decreasing mechanical and cold hypersensitivity induced in a rat model of neuropathic pain. Thus, we compared the effectiveness of pre- or post-treatment with systemic or intrathecal (i.t.) pregabalin at reducing the development and maintenance of the neuropathic pain symptoms. Pregabalin successfully decreased mechanical and cold hypersensitivity, as a pre-treatment, but was less effective at suppressing cold hypersensitivity when administered as a post-treatment. Furthermore, both i.t. and systemic administration of pregabalin were effective in reducing the behavioral hypersensitivity, with the exception of systemic post-treatment on cold hypersensitivity. We also examined pregabalin's effects at inhibiting hind paw formalin-induced nociception in naïve rats and formalin-induced release of excitatory amino acids in the spinal cord dorsal horn (SCDH) both in naïve rats and in rats with neuropathic pain. Pregabalin dose-dependently reduced nociceptive scores in the formalin test. We also present the first evidence that pregabalin reduces the formalin-induced release of glutamate in SCDH. Furthermore, i.t. pregabalin reduces the enhanced noxious stimulus-induced spinal release of glutamate seen in neuropathic rats. These data suggest that pregabalin reduces neuropathic pain symptoms by inhibiting the release of glutamate in the SCDH. Topics: Analgesics; Animals; Chromatography, High Pressure Liquid; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Routes; Drug Administration Schedule; Electrochemistry; gamma-Aminobutyric Acid; Glutamic Acid; Hyperalgesia; Male; Microdialysis; Pain Measurement; Pain Threshold; Physical Stimulation; Pregabalin; Rats; Rats, Long-Evans; Sciatica; Spinal Cord; Spinal Cord Injuries; Statistics, Nonparametric; Time Factors | 2010 |
Interaction of pregabalin with carbamazepine in the mouse maximal electroshock-induced seizure model: a type I isobolographic analysis for non-parallel dose-response relationship curves.
To characterize the anticonvulsant effects of pregabalin (PGB - a third-generation antiepileptic drug) in combination with carbamazepine (CBZ - a classical antiepileptic drug) in the mouse maximal electroshock (MES)-induced seizure model by using the type I isobolographic analysis for non-parallel dose-response relationship curves (DRRCs).. Tonic hind limb extension (seizure activity) was evoked in adult male albino Swiss mice by a current (sine-wave, 25mA, 500V, 50Hz, 0.2s stimulus duration) delivered via auricular electrodes. Potential adverse-effect profiles of interaction of PGB with CBZ at the fixed-ratio of 1:1 in the MES test with respect to motor performance, long-term memory, skeletal muscular strength and antinociceptive activity were measured along with total brain CBZ concentrations.. In the mouse MES model, PGB administered singly had its DRRC non-parallel to that for CBZ. With type I isobolographic analysis for non-parallel DRRCs, the combination of PGB with CBZ at the fixed-ratio of 1:1 exerted additive interaction. In the combination, neither motor coordination, long-term memory nor muscular strength were affected. PGB administered alone and in combination with CBZ exerted antinociceptive effects, whereas CBZ administered alone produced no antinociceptive activity in mice subjected to the acute thermal pain model. Pharmacokinetic estimation of total brain antiepileptic drug concentrations revealed that PGB had no impact on total brain concentrations of CBZ in experimental animals.. In conclusion, the additive interaction between PGB and CBZ is worthy of consideration while extrapolating the results from this study to clinical settings. Topics: Animals; Anticonvulsants; Carbamazepine; Disease Models, Animal; Drug Interactions; Electroshock; gamma-Aminobutyric Acid; Male; Mice; Pregabalin; Seizures | 2010 |
Additive interactions of pregabalin with lamotrigine, oxcarbazepine and topiramate in the mouse maximal electroshock-induced seizure model: a type I isobolographic analysis for non-parallel dose-response relationship curves.
The aim of this study was to characterize the anticonvulsant effects of pregabalin (PGB-a third-generation antiepileptic drug) in combination with three second-generation antiepileptic drugs (i.e., lamotrigine [LTG], oxcarbazepine [OXC] and topiramate [TPM]) in the mouse maximal electroshock (MES)-induced seizure model by using the type I isobolographic analysis for non-parallel dose-response relationship curves (DRRCs). Tonic hind limb extension (seizure activity) was evoked in adult male albino Swiss mice by a current (sine-wave, 25mA, 500V, 50Hz, 0.2s stimulus duration) delivered via auricular electrodes. Potential adverse-effect profiles of interactions of PGB with LTG, OXC and TPM at the fixed-ratio of 1:1 in the MES test with respect to motor performance, long-term memory and skeletal muscular strength were measured. In the mouse MES model, PGB administered singly had its DRRC non-parallel to that for LTG, OXC and TPM. With type I isobolography for non-parallel DRRCs, the combinations of PGB with LTG, OXC and TPM at the fixed-ratio of 1:1 exerted additive interaction. In all combinations, neither motor coordination, long-term memory nor muscular strength were affected. In conclusion, the additive interactions between PGB and LTG, OXC and TPM are worthy of consideration while extrapolating the results from this study to clinical settings. Topics: Animals; Carbamazepine; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Drug Synergism; Drug Therapy, Combination; Electroshock; Fructose; gamma-Aminobutyric Acid; Lamotrigine; Male; Mice; Oxcarbazepine; Pregabalin; Psychomotor Performance; Seizures; Topiramate; Triazines | 2010 |
Oral pregabalin reverses cold allodynia in two distinct models of peripheral neuropathic pain.
A major symptom of persistent neuropathic pain, which may develop after peripheral nerve injury, is hypersensitivity (allodynia) to normally innocuous cold stimuli. Although the anticonvulsant pregabalin has been demonstrated to relieve neuropathic pain, both in preclinical models and clinically, the analgesic effect of the drug in animals has not been profiled for cold hypersensitivity. Therefore, we examined the effect of pregabalin (single oral dosing: 30, 100, 300 micromol/kg) on cold allodynia in two models of chronic neuropathic pain, the spared nerve injury (SNI) and the spinal nerve ligation (SNL) models. A significant antiallodynic effect was observed with pregabalin at all doses tested with a maximal effect of 71% (SNI) and 60% (SNL), respectively compared to vehicle. For comparison, only the highest dose tested of pregabalin (300 micromol/kg), significantly decreased pain responses in phase 2 of the rat formalin test (approximately 67% pain inhibition). However, pregabalin at this high dose also affected other centrally mediated behavioural functions, such as motor activity and anxiolytic behaviour in naïve animals, which could potentially interfere with the pain readout. The present study demonstrates that oral administration of pregabalin significantly reduces both cold allodynia induced in the SNI and the SNL models of neuropathic pain as well as formalin-induced nociception, albeit with different sensitivity and potency. Topics: Administration, Oral; Analgesics; Animals; Behavior, Animal; Chronic Disease; Cold Temperature; Disease Models, Animal; Dose-Response Relationship, Drug; gamma-Aminobutyric Acid; Male; Motor Activity; Pain; Pain Measurement; Peripheral Nervous System Diseases; Pregabalin; Rats; Rats, Sprague-Dawley | 2009 |
The increased trafficking of the calcium channel subunit alpha2delta-1 to presynaptic terminals in neuropathic pain is inhibited by the alpha2delta ligand pregabalin.
Neuropathic pain results from damage to the peripheral sensory nervous system, which may have a number of causes. The calcium channel subunit alpha(2)delta-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of neuropathic pain, and this is causally related to the onset of allodynia, in which a non-noxious stimulus becomes painful. The therapeutic drugs gabapentin and pregabalin (PGB), which are both alpha(2)delta ligands, have antiallodynic effects, but their mechanism of action has remained elusive. To investigate this, we used an in vivo rat model of neuropathy, unilateral lumbar spinal nerve ligation (SNL), to characterize the distribution of alpha(2)delta-1 in DRG neurons, both at the light- and electron-microscopic level. We found that, on the side of the ligation, alpha(2)delta-1 was increased in the endoplasmic reticulum of DRG somata, in intracellular vesicular structures within their axons, and in the plasma membrane of their presynaptic terminals in superficial layers of the dorsal horn. Chronic PGB treatment of SNL animals, at a dose that alleviated allodynia, markedly reduced the elevation of alpha(2)delta-1 in the spinal cord and ascending axon tracts. In contrast, it had no effect on the upregulation of alpha(2)delta-1 mRNA and protein in DRGs. In vitro, PGB reduced plasma membrane expression of alpha(2)delta-1 without affecting endocytosis. We conclude that the antiallodynic effect of PGB in vivo is associated with impaired anterograde trafficking of alpha(2)delta-1, resulting in its decrease in presynaptic terminals, which would reduce neurotransmitter release and spinal sensitization, an important factor in the maintenance of neuropathic pain. Topics: Analysis of Variance; Animals; Anticonvulsants; Behavior, Animal; Calcium Channels; Calcium Channels, L-Type; Disease Models, Animal; Endocytosis; Functional Laterality; gamma-Aminobutyric Acid; Ganglia, Spinal; Male; Microscopy, Electron, Transmission; Neuralgia; Pain Measurement; Pregabalin; Presynaptic Terminals; Protein Transport; Rats; Rats, Sprague-Dawley; Reaction Time; Time Factors; Up-Regulation | 2009 |
Descending serotonergic facilitation and the antinociceptive effects of pregabalin in a rat model of osteoarthritic pain.
Descending facilitation, from the brainstem, promotes spinal neuronal hyperexcitability and behavioural hypersensitivity in many chronic pain states. We have previously demonstrated enhanced descending facilitation onto dorsal horn neurones in a neuropathic pain model, and shown this to enable the analgesic effectiveness of gabapentin. Here we have tested if this hypothesis applies to other pain states by using a combination of approaches in a rat model of osteoarthritis (OA) to ascertain if 1) a role for descending 5HT mediated facilitation exists, and 2) if pregabalin (a newer analogue of gabapentin) is an effective antinociceptive agent in this model. Further, quantitative-PCR experiments were undertaken to analyse the alpha 2 delta-1 and 5-HT3A subunit mRNA levels in L3-6 DRG in order to assess whether changes in these molecular substrates have a bearing on the pharmacological effects of ondansetron and pregabalin in OA.. Osteoarthritis was induced via intra-articular injection of monosodium iodoacetate (MIA) into the knee joint. Control animals were injected with 0.9% saline. Two weeks later in vivo electrophysiology was performed, comparing the effects of spinal ondansetron (10-100 microg/50 microl) or systemic pregabalin (0.3 - 10 mg/kg) on evoked responses of dorsal horn neurones to electrical, mechanical and thermal stimuli in MIA or control rats. In MIA rats, ondansetron significantly inhibited the evoked responses to both innocuous and noxious natural evoked neuronal responses, whereas only inhibition of noxious evoked responses was seen in controls. Pregabalin significantly inhibited neuronal responses in the MIA rats only; this effect was blocked by a pre-administration of spinal ondansetron. Analysis of alpha 2 delta-1 and 5-HT3A subunit mRNA levels in L3-6 DRG revealed a significant increase in alpha 2 delta-1 levels in ipsilateral L3&4 DRG in MIA rats. 5-HT3A subunit mRNA levels were unchanged.. These data suggest descending serotonergic facilitation plays a role in mediating the brush and innocuous mechanical punctate evoked neuronal responses in MIA rats, suggesting an adaptive change in the excitatory serotonergic drive modulating low threshold evoked neuronal responses in MIA-induced OA pain. This alteration in excitatory serotonergic drive, alongside an increase in alpha 2 delta-1 mRNA levels, may underlie pregabalin's state dependent effects in this model of chronic pain. Topics: Analgesics; Animals; Behavior, Animal; Calcium Channels; Calcium Channels, L-Type; Disease Models, Animal; Electrophysiology; gamma-Aminobutyric Acid; Osteoarthritis; Polymerase Chain Reaction; Pregabalin; Rats; Rats, Sprague-Dawley; Serotonin | 2009 |
Brief post-stressor treatment with pregabalin in an animal model for PTSD: short-term anxiolytic effects without long-term anxiogenic effect.
The short- and long-term behavioral effects of a brief course of pregabalin, an antiepileptic structural analogue of alpha-aminobyturic acid with analgesic and anxiolytic effects, were assessed in an animal model of post-traumatic stress disorder (PTSD).. Two-hundred thirty-three adult male Sprague-Dawley rats were employed. Behavioral responses to traumatic stress exposure (predator urine scent) were assessed immediately after (1 h) and 30 days after treatment with saline or pregabalin (at doses of 30, 100 and 300 mg/kg) in terms of behavior in the elevated plus maze (EPM) and the acoustic startle response (ASR) paradigms. At day 31 the freezing response to a trauma cue (clean cat litter) was assessed. The same treatment regimen initiated at day 7 was assessed at day 30 and in response to the trauma cue on day 31 in a separate experiment.. In the short term, doses of 100 mg/kg and 300 mg/kg of pregabalin effectively attenuated anxiety-like behaviors. In the longer-term, pregabalin did not attenuate the onset of PTSD-like behaviors or the prevalence rates of severe cue-responses, for either the immediate or the delayed treatment regimens.. Pregabalin may present an alternative compound for acute anxiolytic treatment after exposure to trauma, but has no long-term protective/preventive effects. Topics: Acoustic Stimulation; Analysis of Variance; Animals; Anticonvulsants; Anxiety; Behavior, Animal; Cues; Disease Models, Animal; Dose-Response Relationship, Drug; Electronic Data Processing; gamma-Aminobutyric Acid; Male; Maze Learning; Pregabalin; Rats; Rats, Sprague-Dawley; Reflex, Acoustic; Stress Disorders, Post-Traumatic; Time Factors | 2008 |
Effects of pregabalin on visceral pain responses and colonic compliance in rats.
Pregabalin, which binds to the alpha2-delta subunit of voltage-gated calcium channels, increased the threshold for pain during colorectal distension (CRD) in irritable bowel syndrome (IBS) patients. We tested the effects of oral pregabalin on the visceral pain-related viscerosomatic and autonomic cardiovascular responses to CRD and colonic compliance in rats.. The activity of the abdominal musculature (viscerosomatic response), monitored by electromyography and intracolonic manometry, and changes in blood pressure and heart rate, monitored by telemetry, were assessed simultaneously in conscious rats during CRD.. Pregabalin (10-200 micromol kg(-1), p.o.) inhibited dose dependently the viscerosomatic response to phasic, noxious CRD (12 distensions at 80 mm Hg). At 200 mumol kg(-1), pregabalin also reduced the increase in blood pressure and heart rate associated with noxious CRD. Moreover, pregabalin (200 micromol kg(-1), p.o.) reduced the visceromotor response to ascending phasic CRD (10-80 mm Hg) and significantly increased the threshold pressure for response. During phasic CRD (2-20 mm Hg), pregabalin (200 micromol kg(-1), p.o.) increased intracolonic volume, resulting in a shift to the left of the pressure-volume relationship curve, indicative of an increase of compliance.. Pregabalin reduced the viscerosomatic and autonomic responses associated with CRD-induced visceral pain and increased colonic compliance in rats. These observations confirm the analgesic activity of pregabalin on visceral pain and support the translational value of the CRD model to humans. Ligands for the alpha2-delta subunit might represent interesting compounds for the treatment of visceral pain disorders, such as IBS. Topics: Abdominal Pain; Administration, Oral; Analgesics; Animals; Blood Pressure; Calcium Channels; Colon; Compliance; Dilatation, Pathologic; Disease Models, Animal; Dose-Response Relationship, Drug; Female; gamma-Aminobutyric Acid; Heart Rate; Irritable Bowel Syndrome; Manometry; Pregabalin; Protein Subunits; Rats; Rats, Sprague-Dawley | 2008 |
Comparative antiallodynic activity of morphine, pregabalin and lidocaine in a rat model of neuropathic pain produced by one oxaliplatin injection.
A single infusion of oxaliplatin, a drug active against colorectal cancer, induces specific painful syndrome characterized by neurosensitive symptoms triggered or aggravated in cold conditions. In an animal model that reproduces such hypersensitivity to cold for five days after a single oxaliplatin administration (6mg/kg, i.p.), we assessed the antinociceptive efficacy of intravenously administered drugs such as morphine, lidocaine and pregabalin using the rat tail immersion test in cold water (10 degrees C). The antinociceptive efficacy was first ranked by ratio of the pharmacological effect (versus time) to dose: pregabalin (2mg/kg)>lidocaine (3mg/kg)>morphine (4mg/kg). Our results show that pregabalin may be a good choice to treat cold hypersensitivity after one oxaliplatin injection. Topics: Analgesics; Analysis of Variance; Animals; Behavior, Animal; Disease Models, Animal; Dose-Response Relationship, Drug; gamma-Aminobutyric Acid; Lidocaine; Male; Morphine; Neuralgia; Organoplatinum Compounds; Oxaliplatin; Pain Measurement; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Reaction Time | 2008 |
Pharmacologic investigation of the mechanism underlying cold allodynia using a new cold plate procedure in rats with chronic constriction injuries.
Cold allodynia is a frequent clinical symptom of patients with neuropathic pain. Despite numerous studies of cold allodynia, using animal models of neuropathic pain, little is known about its underlying mechanisms. This study was performed to establish a method for the pharmacologic evaluation of cold allodynia using several analgesics in a chronic constriction injury (CCI) rat model of neuropathic pain. Compared with the results obtained before the CCI operation, the CCI rats placed on a cork plate at 20 degrees C exhibited a slight change in the paw withdrawal latency because of the mechanical stimulus mediated by the injured paw touching the plate. By contrast, there was a significant reduction in the paw withdrawal latency on a cold metal plate compared with that on the cork plate after the CCI surgery, with the maximum decrease occurring on postoperative day 7. This reduction is thought to specifically reflect cold-induced pain behavior. In addition, both naïve and CCI rats showed behavioral changes at 5 and 0 degrees C, but not at 10 degrees C or higher. Interestingly, a subcutaneous morphine dose of 6 mg/kg completely inhibited cold allodynia induced at 10 degrees C on postoperative day 7. Under this condition, both the sodium channel blocker mexiletine (10 and 30 mg/kg, subcutaneously) and the calcium channel alpha2delta subunit blocker pregabalin (30 and 100 mg/kg, orally) significantly suppressed cold allodynia. Additionally, both resiniferatoxin (0.3 mg/kg, subcutaneously), an ultrapotent analog of capsaicin that desensitizes C fibers, and the VR1 channel antagonist N-(4-tertiarybutylphenyl)-4-(3-chloropyridin-2-yl) tetrahydropyrazine-1(2H)-carboxamide (10 and 30 mg/kg, orally) significantly prolonged the paw withdrawal latency. In conclusion, our data suggest that the activation of C fibers mediates cold allodynia. Topics: Analgesics, Opioid; Animals; Behavior, Animal; Calcium Channel Blockers; Chronic Disease; Cold Temperature; Constriction, Pathologic; Disease Models, Animal; Diterpenes; gamma-Aminobutyric Acid; Male; Mexiletine; Morphine; Nerve Fibers, Unmyelinated; Pain; Pain Measurement; Peripheral Nervous System Diseases; Pregabalin; Pyrazines; Pyridines; Rats; Rats, Sprague-Dawley; Reaction Time; Sciatic Nerve; Sodium Channel Blockers; TRPV Cation Channels | 2008 |
Pregabalin reduces muscle and cutaneous hyperalgesia in two models of chronic muscle pain in rats.
Pregabalin is used for treatment of neuropathic pain conditions. The present study evaluated effects of pregabalin in 2 rat models of muscle-induced hyperalgesia: Inflammatory and noninflammatory. Muscle hyperalgesia (withdrawal threshold to compression of the muscle) and cutaneous hyperalgesia of the paw (withdrawal threshold to von Frey filaments) were measured before and after induction of hyperalgesia and after treatment with pregabalin (saline, 10 to 100 mg/kg i.p.). In the inflammatory model, 3% carrageenan injected into 1 gastrocnemius muscle decreased the mechanical withdrawal threshold of the paw bilaterally and the compression withdrawal threshold of the muscle ipsilaterally 2 weeks later. Pregabalin (10 to 100 mg/kg) increased the compression withdrawal threshold of the inflamed muscle when compared with vehicle controls. Pregabalin also increased the mechanical withdrawal threshold of the paw bilaterally, but only with 100 mg/kg. In the noninflammatory model, 2 unilateral injections of acidic saline into the gastrocnemius muscle produced bilateral cutaneous and muscle hyperalgesia 24 hours after the second injection. Pregabalin (10 to 100 mg/kg i.p.) significantly increased the compression withdrawal thresholds of the muscle and the mechanical withdrawal threshold of the paw bilaterally when compared with vehicle. However, pregabalin also has significant motor effects at the higher doses (60 to 100 mg/kg). Therefore, pregabalin reduces both muscle and cutaneous hyperalgesia that occurs after muscle insult in 2 animal models of muscle pain at doses that do not produce ataxia.. This study shows that pregabalin reduces both cutaneous and muscle hyperalgesia in inflammatory and noninflammatory models of muscle pain. Thus, pregabalin may be an effective treatment for people with chronic muscle pain. Topics: Analgesics; Animals; Carrageenan; Chronic Disease; Disease Models, Animal; Dose-Response Relationship, Drug; Functional Laterality; gamma-Aminobutyric Acid; Hyperalgesia; Male; Motor Activity; Muscle, Skeletal; Muscular Diseases; Pain; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Reaction Time; Skin; Time Factors | 2007 |
Antiallodynic effect of pregabalin in rat models of sympathetically maintained and sympathetic independent neuropathic pain.
Pregabalin binds to the voltage-dependent calcium channel alpha2delta subunit and modulates the release of neurotransmitters, resulting in analgesic effects on neuropathic pain. Neuropathic pain has both sympathetically maintained pain (SMP) and sympathetic independent pain (SIP) components. We studied the antiallodynic effects of pregabalin on tactile allodynia (TA) and cold allodynia (CA) in SMP-and SIP-dominant neuropathic pain models. Allodynia was induced by ligation of the L5 and L6 spinal nerves (SMP model) or by transection of the tibial and sural nerves (SIP model) in rats. For intrathecal drug administration, a PE-10 catheter was implanted through the atlantooccipital membrane to the lumbar enlargement. Pregabalin was administered either intraperitoneally (IP) or intrathecally (IT) and dosed up incrementally until an antiallodynic effect without sedation or motor impairment was apparent. TA was assessed using von Frey filaments, and CA was assessed using acetone drops. IP-administered pregabalin dose-dependently attenuated TA in both models and CA in the SMP model, but not CA in the SIP model. IT-administered pregabalin dose-dependently attenuated both TA and CA in both models. However, the dose response curve of IT-administered pregabalin in SMP was shifted to left from that of SIP and the ED50 of IT-administered pregabalin for CA in SMP was about 900 times less than that in SIP. These findings suggest that pregabalin exerts its antiallodynic effect mainly by acting at the spinal cord, and that IT-administered pregabalin has more potent antiallodynic effects in SMP. The alpha2delta subunit might be less involved in the CA in SIP. Topics: Analgesics; Animals; Disease Models, Animal; Dose-Response Relationship, Drug; gamma-Aminobutyric Acid; Male; Neuralgia; Pain Measurement; Pain Threshold; Pregabalin; Rats; Rats, Sprague-Dawley; Sympathetic Nervous System | 2007 |
Pregabalin, S-(+)-3-isobutylgaba, activates the descending noradrenergic system to alleviate neuropathic pain in the mouse partial sciatic nerve ligation model.
We have previously demonstrated that gabapentin supraspinally activates the descending noradrenergic system to alleviate neuropathic pain. In this study, we investigated whether pregabalin, an antiepileptic and analgesic drug that is also designed as a structural analogue of gamma-aminobutyric acid (GABA), exhibits supraspinal analgesic effects similar to those of gabapentin involving the descending noradrenergic system. Both systemically (intraperitoneally; i.p.) and locally (intracerebroventricularly or intrathecally; i.c.v. or i.t.) injected pregabalin reduced thermal and mechanical hypersensitivity in a murine chronic pain model that was prepared by partial ligation of the sciatic nerve (the Seltzer model), suggesting that pregabalin acts at both supraspinal and spinal loci. The supraspinal analgesic action of pregabalin was observed only after peripheral nerve injury, and pregabalin (i.p. and i.c.v.) did not affect acute thermal and mechanical nociception. Depletion of spinal noradrenaline (NA) or pharmacological blockade of spinal alpha(2)-adrenoceptors with yohimbine (i.p. or i.t.), but not alpha(1)-adrenoceptors with prazosin (i.p.), reduced the analgesic effects of pregabalin (i.p. or i.c.v.) on thermal and mechanical hypersensitivity. Moreover, i.c.v.-administered pregabalin dose-dependently increased the spinal 4-hydroxy-3-methoxyphenylglycol (MHPG) content and the MHPG/NA ratio only in mice with neuropathic pain, whereas the concentrations of NA, serotonin, 5-hydroxyindoleacetic acid and dopamine were unchanged, demonstrating that supraspinal pregabalin accelerated the spinal turnover of NA. Together, these results indicate that pregabalin supraspinally activates the descending noradrenergic pain inhibitory system coupled with spinal alpha(2)-adrenoceptors to ameliorate neuropathic pain. Topics: Adrenergic Agents; Afferent Pathways; Analgesics; Animals; Behavior, Animal; Brain Stem; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Routes; gamma-Aminobutyric Acid; Hyperalgesia; Locomotion; Male; Methoxyhydroxyphenylglycol; Mice; Norepinephrine; Oxidopamine; Pain Measurement; Pregabalin; Sciatic Neuropathy; Spinal Cord; Time Factors; Yohimbine | 2007 |
Heteroaromatic side-chain analogs of pregabalin.
A series of heteroaromatic analogs of pregabalin has been identified that possess anticonvulsant activity in the DBA/2 mouse model. The methods of synthesis and preliminary pharmacology are discussed herein. Topics: Animals; Anticonvulsants; Disease Models, Animal; Drug Design; Drug Evaluation, Preclinical; gamma-Aminobutyric Acid; In Vitro Techniques; Mice; Mice, Inbred DBA; Molecular Structure; Pregabalin; Seizures; Stereoisomerism | 2006 |
Carboxylate bioisosteres of pregabalin.
Several beta-amino tetrazole analogs of gabapentin 1 and pregabalin 2 were prepared by one of two convergent, highly efficient routes, and their affinity for the alpha(2)-delta protein examined. Two select compounds with potent affinity for alpha(2)-delta, 8a and 16a, were subsequently tested in vivo in an audiogenic seizure model and found to elicit protective effects. Topics: Amines; Animals; Anticonvulsants; Binding Sites; Carboxylic Acids; Cyclohexanecarboxylic Acids; Disease Models, Animal; Drug Evaluation, Preclinical; Epilepsy, Reflex; Gabapentin; gamma-Aminobutyric Acid; Mice; Mice, Inbred DBA; Molecular Structure; Pregabalin; Protein Subunits; Stereoisomerism; Structure-Activity Relationship | 2006 |
Activity profile of pregabalin in rodent models of epilepsy and ataxia.
Pregabalin (Lyrica) is a novel amino acid compound that binds with high affinity to the alpha2-delta (alpha2-delta) auxiliary protein of voltage-gated calcium channels. In vivo, it potently prevents seizures, pain-related behaviors and has anxiolytic-like activity in rodent models. The present studies were performed to determine the profile of pregabalin anticonvulsant activity in a variety of mouse and rat models. In the high-intensity electroshock test, pregabalin potently inhibited tonic extensor seizures in rats (ED50 = 1.8 mg/kg, PO), and low-intensity electroshock seizures in mice. It prevented tonic extensor seizures in the DBA/2 audiogenic mouse model (ED50 = 2.7 mg/kg, PO). Its time course of action against electroshock induced seizures in rats roughly followed the pharmacokinetics of radiolabeled drug in the brain compartment. At higher dosages (ED50 1= 31 mg/kg, PO), pregabalin prevented clonic seizures from pentylenetetrazole in mice. In a kindled rat model of partial seizures, pregabalin prevented stages 4-5 behavioral seizures (lowest effective dose = 10 mg/kg, IP), and also reduced the duration of electrographic seizures. Pregabalin was not active to prevent spontaneous absence-like seizures in the Genetic Absence Epilepsy in Rats from Strasbourg (GAERS) inbred Wistar rat strain. Pregabalin caused ataxia and decreased spontaneous locomotor activity at dosages 10-30-fold higher than those active to prevent seizures. These findings suggest that pregabalin has an anticonvulsant mechanism different from the prototype antiepileptic drugs and similar to that of gabapentin except with increased potency and bioavailability. In summary, our results show that pregabalin has several properties that favor treatment of partial seizures in humans. Topics: Animals; Anticonvulsants; Behavior, Animal; Disease Models, Animal; Dose-Response Relationship, Drug; Epilepsy; Female; Gait Ataxia; gamma-Aminobutyric Acid; Kindling, Neurologic; Male; Mice; Mice, Inbred Strains; Motor Activity; Pregabalin; Rats; Rats, Sprague-Dawley; Rats, Wistar; Seizures | 2006 |
Oral treatment with PD-0200347, an alpha2delta ligand, reduces the development of experimental osteoarthritis by inhibiting metalloproteinases and inducible nitric oxide synthase gene expression and synthesis in cartilage chondrocytes.
To examine the in vivo effects of PD-0200347, an alpha(2)delta ligand of voltage-activated Ca(2+) channels and a compound chemically related to pregabalin and gabapentin, on the development of cartilage structural changes in an experimental dog model of osteoarthritis (OA). The effects of PD-0200347 on the major pathways involved in OA cartilage degradation, including matrix metalloproteinases (MMPs) and the inducible form of nitric oxide synthase (iNOS), were also studied.. OA was surgically induced in dogs by sectioning the anterior cruciate ligament. OA dogs were randomly distributed into 3 groups and treated orally with either 1) placebo, 2) 15 mg/kg/day of PD-0200347, or 3) 90 mg/kg/day of PD-0200347. Dogs were killed 12 weeks after surgery. The severity of the lesions was scored macroscopically and histologically. Cartilage specimens from the femoral condyles and tibial plateaus were processed for RNA extraction and quantitative reverse transcription-polymerase chain reaction (RT-PCR) or immunohistochemistry. Specific probes and antibodies were used to study the messenger RNA and protein levels of iNOS, MMP-1, MMP-3, and MMP-13.. No clinical signs of drug toxicity were noted in the treated animals. Treatment with PD-0200347 at both dosages tested (15 and 90 mg/kg/day) reduced the development of cartilage lesions. There was a reduction in the score of lesions, with a statistically significant (P = 0.01) difference when the highest dosage of the drug was administered. The reduction in the score was mainly related to a decrease in the surface size of the lesions. Quantitative RT-PCR showed that PD-0200347 significantly reduced the expression of MMP-13, a key mediator in OA. Immunohistochemical analyses showed that treatment with PD-0200347 significantly reduced the synthesis of all key OA mediators studied.. This study demonstrated the efficacy of PD-0200347 in reducing the progression of cartilage structural changes in a dog model of OA. It also showed that this effect is linked to the inhibition of the major pathophysiologic mediators responsible for cartilage degradation. Topics: Administration, Oral; Amines; Animals; Cartilage, Articular; Chondrocytes; Cyclohexanecarboxylic Acids; Disease Models, Animal; Disease Progression; Dogs; Gabapentin; gamma-Aminobutyric Acid; Gene Expression; Immunohistochemistry; Metalloproteases; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Osteoarthritis; Pregabalin; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger | 2005 |
Design, synthesis, and preliminary evaluation of gabapentin-pregabalin mutual prodrugs in relieving neuropathic pain.
As a part of a program for the development of specific analgesics in relieving neuropathic pain, the purpose of the present study was to investigate a new concept that involves the conjugation of two drugs, gabapentin and pregabalin, as mutual prodrugs using a chemical modification approach. A series of gabapentin-pregabalin diester compounds were synthesized using linear or branch bis-hydroxyl linkers. Their pharmacological properties for treating neuropathic pain were investigated in a rat model of chronic sciatic nerve constriction injury (CCI). In-vivo evaluation demonstrated that 1a and 1b composed of two gabapentin molecules as well as 3a composed of gabapentin and pregabalin with the short linear linker, were effective in reversing tactile allodynia in CCI rats. Compounds with longer or side-branched linkers showed lower efficiencies and severe adverse effects. Topics: Amines; Analgesics, Non-Narcotic; Animals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Drug Design; Gabapentin; gamma-Aminobutyric Acid; Male; Molecular Structure; Pregabalin; Prodrugs; Rats; Rats, Sprague-Dawley; Sciatic Neuropathy; Structure-Activity Relationship | 2005 |
Effect of gabapentin derivates on mechanical allodynia-like behaviour in a rat model of chronic sciatic constriction injury.
A series of mutual prodrugs derived from gabapentin, pregabalin, memantine, venlafaxine were synthesized and their pharmacological properties to treat neuropathic pain were investigated in a rat model of chronic sciatic nerve constriction injury (CCI). In vivo evaluation demonstrated that the mutual prodrugs 2002413A, 2002823A composed of two gabapentins, 2002414 composed of gabapentin and pregabalin were effective in reversal tactile allodynia in CCI rats. The prodrugs 2002413A, 2002414 had no significant influence on the rotarod activity. The result suggest that the prodrugs may be possible candidates for further development. Topics: Amines; Animals; Chronic Disease; Cyclohexanecarboxylic Acids; Cyclohexanols; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Memantine; Pain; Pain Threshold; Pregabalin; Prodrugs; Rats; Sciatic Neuropathy; Structure-Activity Relationship; Venlafaxine Hydrochloride | 2004 |
The biology and pharmacology of calcium channel alpha2-delta proteins Pfizer Satellite Symposium to the 2003 Society for Neuroscience Meeting. Sheraton New Orleans Hotel, New Orleans, LA November 10, 2003.
Topics: Acetates; Amines; Analgesics; Animals; Anticonvulsants; Binding Sites; Calcium Channels; Calcium Channels, L-Type; Cyclohexanecarboxylic Acids; Disease Models, Animal; Gabapentin; gamma-Aminobutyric Acid; Humans; Mice; Pregabalin; Rats; Societies, Scientific | 2004 |
Future Pain Drugs - Europe 2003. 15-16 September 2003, London, UK.
Topics: Acetaminophen; Acetates; Amines; Analgesics; Animals; Cyclohexanecarboxylic Acids; Disease Models, Animal; Drug Delivery Systems; Fructose; Gabapentin; gamma-Aminobutyric Acid; Humans; Levetiracetam; Nitrates; Pain; Peripheral Nervous System Diseases; Piracetam; Pregabalin; Topiramate | 2003 |