Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Pharm Compd ; 28(2): 100-109, 2024.
Article in English | MEDLINE | ID: mdl-38604146

ABSTRACT

Pain syndromes are among the most widespread, costly, and debilitating of all neurological disorders. The number of patients living with chronic pain is expected to increase with the aging population and with the rise in obesity and diabetes across the nation. This type of pain is often insensitive to the traditional pain pharmacopeia or surgical intervention. Over the last 10 years the number of prescriptions that have been compounded by pharmacists has increased dramatically. There are a number of drugs in the area of pain management that have been formulated and compounded by pharmacists to treat conditions such as diabetic neuropathy, fibromyalgia, postherpetic neuralgia, joint pain, arthritis, and a variety of other conditions. A significant portion of these compounded analgesic preparations is made up of topical/transdermal dosage forms such as gels and creams. While the efficacy and doses of these drugs in systemic dosage forms have been widely established, little is known about the permeation and efficacy of these compounds from topical/transdermal gels. This review will provide an overview of chronic pain as a disease, the mechanisms of chronic pain, current treatment approaches to chronic pain, and a discussion of the drugs that are typically compounded into these topical formulations and studied in clinical trials.


Subject(s)
Chronic Pain , Neuralgia, Postherpetic , Neuralgia , Humans , Analgesics , Chronic Pain/drug therapy , Gels/therapeutic use , Neuralgia/drug therapy , Neuralgia, Postherpetic/drug therapy
2.
J Pain ; : 104508, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38484854

ABSTRACT

Postamputation pain is currently managed unsatisfactorily with neuron-targeted pharmacological and interventional therapies. Non-neuronal pain mechanisms have emerged as crucial factors in the development and persistence of postamputation pain. Consequently, these mechanisms offer exciting prospects as innovative therapeutic targets. We examined the hypothesis that engaging mesenchymal stem cells (MSCs) would foster local neuroimmune interactions, leading to a potential reduction in postamputation pain. We utilized an ex vivo neuroma model from a phantom limb pain patient to uncover that the oligodeoxynucleotide IMT504 engaged human primary MSCs to promote an anti-inflammatory microenvironment. Reverse translation experiments recapitulated these effects. Thus, in an in vivo rat model, IMT504 exhibited strong efficacy in preventing autotomy (self-mutilation) behaviors. This effect was linked to a substantial accumulation of MSCs in the neuroma and associated dorsal root ganglia and the establishment of an anti-inflammatory phenotype in these compartments. Centrally, this intervention reduced glial reactivity in the dorsal horn spinal cord, demonstrating diminished nociceptive activity. Accordingly, the exogenous systemic administration of MSCs phenocopied the behavioral effects of IMT504. Our findings underscore the mechanistic relevance of MSCs and the translational therapeutic potential of IMT504 to engage non-neuronal cells for the prevention of postamputation pain. PERSPECTIVE: The present study suggests that IMT504-dependent recruitment of endogenous MSCs within severely injured nerves may prevent post-amputation pain by modifying the inflammatory scenario at relevant sites in the pain pathway. Reinforcing data in rat and human tissues supports the potential therapeutic value of IMT504 in patients suffering postamputation pain.

3.
Mol Pain ; 13: 1744806916688220, 2017 01.
Article in English | MEDLINE | ID: mdl-28326930

ABSTRACT

Background JWH015 is a cannabinoid (CB) receptor type 2 agonist that produces immunomodulatory effects. Since skin cells play a key role in inflammatory conditions and tissue repair, we investigated the ability of JWH015 to promote an anti-inflammatory and pro-wound healing phenotype in human primary skin cells. Methods Human primary keratinocytes and fibroblasts were stimulated with lipopolysaccharide. The mRNA expression of cannabinoid receptors was determined using RT-PCR. The effects of JWH015 (0.05, 0.1, 0.5, and 1 µM) in pro- and anti-inflammatory factors were tested in lipopolysaccharide-stimulated cells. A scratch assay, using a co-culture of keratinocytes and fibroblasts, was used to test the effects of JWH015 in wound healing. In addition, the topical and transdermal penetration of JWH015 was studied in Franz diffusion cells using porcine skin and LC-MS. Results The expression of CB1 and CB2 receptors (mRNA) and the production of pro- and anti-inflammatory factors enhanced in keratinocytes and fibroblasts following lipopolysaccharide stimulation. JWH015 reduced the concentration of major pro-inflammatory factors (IL-6 and MCP-1) and increased the concentration of a major anti-inflammatory factor (TGF-ß) in lipopolysaccharide-stimulated cells. JWH015 induced a faster scratch gap closure. These JWH015'seffects were mainly modulated through both CB1 and CB2 receptors. Topically administered JWH015 was mostly retained in the skin and displayed a sustained and low level of transdermal permeation. Conclusions Our findings suggest that targeting keratinocytes and fibroblasts with cannabinoid drugs could represent a therapeutic strategy to resolve peripheral inflammation and promote tissue repair.


Subject(s)
Cannabinoid Receptor Agonists/administration & dosage , Cytokines/metabolism , Fibroblasts/drug effects , Indoles/pharmacology , Keratinocytes/drug effects , Administration, Cutaneous , Cannabinoid Receptor Antagonists/pharmacology , Cells, Cultured , Cytokines/genetics , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Lipopolysaccharides/pharmacology , Mass Spectrometry , RNA, Messenger/metabolism , Time Factors , Wound Healing/drug effects
4.
J Pain ; 17(6): 654-68, 2016 06.
Article in English | MEDLINE | ID: mdl-26961090

ABSTRACT

UNLABELLED: Cannabinoids show promise as therapeutic agents, particularly as analgesics, but their development and clinical use has been complicated by recognition of their botanical source, cannabis, as a substance of misuse. Although research into endogenous cannabinoid systems and potential cannabinoid pharmaceuticals is slowly increasing, there has been intense societal interest in making herbal (plant) cannabis available for medicinal use; 23 U.S. States and all Canadian provinces currently permit use in some clinical contexts. Whether or not individual professionals support the clinical use of herbal cannabis, all clinicians will encounter patients who elect to use it and therefore need to be prepared to advise them on cannabis-related clinical issues despite limited evidence to guide care. Expanded research on cannabis is needed to better determine the individual and public health effects of increasing use of herbal cannabis and to advance understanding of the pharmaceutical potential of cannabinoids as medications. This article reviews clinical, research, and policy issues related to herbal cannabis to support clinicians in thoughtfully advising and caring for patients who use cannabis, and it examines obstacles and opportunities to expand research on the health effects of herbal cannabis and cannabinoids. PERSPECTIVE: Herbal cannabis is increasingly available for clinical use in the United States despite continuing controversies over its efficacy and safety. This article explores important considerations in the use of plant Cannabis to better prepare clinicians to care for patients who use it, and identifies needed directions for research.


Subject(s)
Cannabinoids/therapeutic use , Pain Management/methods , Pain/drug therapy , Animals , Biomedical Research , Clinical Trials as Topic , Humans , Pain/physiopathology , United States
5.
Int J Pharm Compd ; 19(2): 167-73, 2015.
Article in English | MEDLINE | ID: mdl-26685497

ABSTRACT

Many patients with chronic neuropathic pain continue to suffer despite traditional pharmacotherapy. As a result, pharmacists commonly compound gabapentin into creams, gels, and ointments as an alternative treatment option. In this study, various concentrations (1%, 5%, and 10%) of topical gabapentin compounded in Lipoderm were applied at various pre-treatment times (30 minutes, 1 hour, and 4 hours) to investigate what gabapentin concentration and pre-treatment time best attenuates formalin-induced nociceptive behaviors in a rodent model. A 30-minute pre-treatment with 5% gabapentin demonstrated maximum attenuation of nociceptive behaviors in this in vivo preclinical pain model. Nociceptive behaviors unexpectedly increased when gabapentin concentration or pre-treatment time was increased, suggesting both antinociceptive and pronociceptive effects of transdermal gabapentin administration. Gabapentin permeation into the skin and deeper tissues of the hindpaw was measured following the in vivo study. Skin and deep tissue permeation of gabapentin was both dose and time-dependent. Maximum deep-tissue permeation occurred within 30 minutes of topical application. Skin concentrations increased with a longer 1-hour pre-treatment. Minimal skin and deeper tissue levels were found following a 4-hour pre-treatment. These results suggest that topical gabapentin may be antinociceptive in a rodent formalin model at specific doses and pre-treatment intervals.


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Skin/metabolism , gamma-Aminobutyric Acid/pharmacology , Administration, Topical , Amines/pharmacokinetics , Animals , Cricetinae , Cyclohexanecarboxylic Acids/pharmacokinetics , Gabapentin , Male , Mesocricetus , Ointments , gamma-Aminobutyric Acid/pharmacokinetics
6.
Int J Pharm Compd ; 18(4): 278-86, 2014.
Article in English | MEDLINE | ID: mdl-25474856

ABSTRACT

Pain syndromes are among the most widespread, costly, and debilitating of all neurological disorders. The number of patients living with chronic pain is expected to increase with the aging population and with the rise in obesity and diabetes across the nation. This type of pain is often insensitive to the traditional pain pharmacopeia or surgical intervention. Over the last 10 years the number of prescriptions that have been compounded by pharmacists has increased dramatically. There are a number of drugs in the area of pain management that have been formulated and compounded by pharmacists to treat conditions such as diabetic neuropathy, fibromyalgia, postherpetic neuralgia, joint pain, arthritis, and a variety of other conditions. A significant portion of these compounded analgesic preparations is made up of topical/transdermal dosage forms such as gels and creams. While the efficacy and doses of these drugs in systemic dosage forms have been widely established, little is known about the permeation and efficacy of these compounds from topical/transdermal gels. This review will provide an overview of chronic pain as a disease, the mechanisms of chronic pain, current treatment approaches to chronic pain, and a discussion of the drugs that are typically compounded into these topical formulations and studied in clinical trials.


Subject(s)
Analgesics/administration & dosage , Pain/drug therapy , Administration, Topical , Analgesics/chemistry , Analgesics/therapeutic use , Drug Compounding , Humans , Pain/physiopathology
7.
Pain ; 150(3): 401-413, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573450

ABSTRACT

Anti-nociceptive tolerance to opioids is a well-described phenomenon, which severely limits the clinical efficacy of opioids for the treatment of chronic pain syndromes. The mechanisms that drive anti-nociceptive tolerance, however, are less well understood. We have previously shown that glia have a central role in the development of morphine tolerance and that administration of a glial modulating agent attenuated tolerance formation. Recently, we have demonstrated that morphine enhances microglial Iba1 expression and P2X4 receptor-mediated microglial migration via direct mu opioid receptor signaling in in vitro microglial cultures. We hypothesize that P2X4 receptors drive morphine tolerance and modulate morphine-induced spinal glial reactivity. Additionally, we hypothesize that perivascular microglia play a role in morphine tolerance and that P2X4 receptor expression regulates perivascular microglia ED2 expression. To test these hypotheses, rats were implanted with osmotic minipumps releasing morphine or saline subcutaneously for seven days. Beginning three days prior to morphine treatment, P2X4 receptor antisense oligonucleotide (asODN) was injected intrathecally daily, to selectively inhibit P2X4 receptor expression. P2X4 receptor asODN treatment inhibited morphine-induced P2X4 receptor expression and blocked anti-nociceptive tolerance to systemically administered morphine. P2X4 receptor asODN treatment also attenuated the morphine-dependent increase of spinal ionized calcium binding protein (Iba1), glial fibrillary acidic protein (GFAP) and mu opioid receptor protein expression. Chronic morphine also decreased perivascular microglial ED2 expression, which was reversed by P2X4 receptor asODN. Together, these data suggest that the modulation of P2X4 receptor expression on microglia and perivascular microglia may prove an attractive target for adjuvant therapy to attenuate opioid-induced anti-nociceptive tolerance.


Subject(s)
Calcium-Binding Proteins/metabolism , Drug Tolerance/physiology , Gene Expression Regulation/physiology , Microglia/metabolism , Morphine/administration & dosage , Receptors, Opioid/metabolism , Receptors, Purinergic P2X4/metabolism , Analysis of Variance , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Behavior, Animal/drug effects , CD11b Antigen/metabolism , Disease Models, Animal , Drug Interactions , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Male , Microfilament Proteins , Microglia/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Pain Measurement , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Receptors, Purinergic P2X4/genetics , Signal Transduction , Spinal Cord/pathology , Time Factors
8.
Brain Res ; 1219: 116-26, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18538310

ABSTRACT

The treatment of acute and chronic pain is still deficient. The modulation of glial cells may provide novel targets to treat pain. We hypothesize that astrocytes and microglia participate in the initiation and maintenance of both, acute surgical and chronic neuropathic pain. Rats underwent paw incision, L5 nerve exposure or L5 nerve transection surgery. Behavioral mechanical allodynia was assessed using von Frey filaments. Immunohistochemistry was performed using anti-ionized calcium binding adaptor protein, Iba-1 (microglia), and anti-Glial Fibrillary Acidic Protein, GFAP (astrocytes) on day 1, 4 and 7 after surgery. Following paw incision and at spinal L5 segment GFAP expression was increased in laminae I-II and Iba1 in deep laminae on day 1, in the entire dorsal horn on day 4 and dissipated on day 7 after paw incision in parallel with the allodynia. L5 nerve transection induced mechanical allodynia from day 1 to 7 which correlated with Iba-1 increases on day 1, 4 (entire dorsal horn) and day 7 after nerve injury (deep laminae of the dorsal horn) at spinal L5 segment. Conversely, GFAP increased at later time points from day 4 (deep laminae) and on day 7 (entire dorsal horn). Our data demonstrates that astrocytes (GFAP expression) play a role in the initiation of acute pain and the maintenance of chronic pain while Iba-1 increases closely correlated with the early phase of neuropathic pain. Iba1 and GFAP increased rostrally, at L3 segment, after paw incision (day 4) and only Iba1 increased following L5 nerve transection (day 7).


Subject(s)
Calcium-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Pain/pathology , Spinal Cord/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Functional Laterality , Male , Microfilament Proteins , Neuroglia/metabolism , Pain Measurement , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Nerves/injuries , Time Factors
9.
Anesthesiology ; 108(4): 722-34, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18362605

ABSTRACT

BACKGROUND: Cannabinoids induce analgesia by acting on cannabinoid receptor (CBR) types 1 and/or 2. However, central nervous system side effects and antinociceptive tolerance from CBR1 limit their clinical use. CBR2 exist on spinal glia and perivascular cells, suggesting an immunoregulatory role of these receptors in the central nervous system. Previously, the authors showed that spinal CBR2 activation reduces paw incision hypersensitivity and glial activation. This study tested whether CBR2 are expressed in glia and whether their activation would induce antinociception, glial inhibition, central side effects, and antinociceptive tolerance in a neuropathic rodent pain model. METHODS: Rats underwent L5 spinal nerve transection or sham surgery, and CBR2 expression and cell localization were assessed by immunohistochemistry. Animals received intrathecal injections of CBR agonists and antagonists, and mechanical withdrawal thresholds and behavioral side effects were assessed. RESULTS: Peripheral nerve transection induced hypersensitivity, increased expression of CR3/CD11b and CBR2, and reduced ED2/CD163 expression in the spinal cord. The CBR2 were localized to microglia and perivascular cells. Intrathecal JWH015 reduced peripheral nerve injury hypersensitivity and CR3/CD11b expression and increased ED2/CD163 expression in a dose-dependent fashion. These effects were prevented by intrathecal administration of the CBR2 antagonist (AM630) but not the CBR1 antagonist (AM281). JWH015 did not cause behavioral side effects. Chronic intrathecal JWH015 treatment did not induce antinociceptive tolerance. CONCLUSIONS: These data indicate that intrathecal CBR2 agonists may provide analgesia by modulating the spinal immune response and microglial function in chronic pain conditions without inducing tolerance and neurologic side effects.


Subject(s)
Drug Tolerance/physiology , Microglia/metabolism , Oligodendroglia/metabolism , Peripheral Nerve Injuries , Receptor, Cannabinoid, CB2/biosynthesis , Spinal Cord/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cyclohexanols/pharmacology , Cyclohexanols/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Male , Microglia/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Pain Measurement/drug effects , Pain Measurement/methods , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists , Spinal Cord/cytology , Spinal Cord/drug effects
10.
Anesthesiology ; 106(4): 787-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17413917

ABSTRACT

BACKGROUND: Cannabinoids bind to cannabinoid receptors type 1 and 2 and produce analgesia in several pain models, but central side effects from cannabinoid 1 receptors limit their clinical use. Cannabinoid 2 receptors reduce inflammatory responses in the periphery by acting on immune cells, and they are present on glia in the central nervous system. This study tested whether spinal cannabinoid activation would induce analgesia, glial inhibition, and central side effects in a postoperative model or incisional pain. METHODS: Rats underwent paw incision surgery, with intrathecal injections of cannabinoid agonists and antagonists and assessment of withdrawal thresholds and behavioral side effects. Spinal glial activation was determined by immunohistochemistry. RESULTS: Intrathecal administration CP55940 reduced postoperative hypersensitivity (91 +/- 9% maximum possible effect; P < 0.05), and this was prevented by intrathecal administration of both cannabinoid 1 receptor (AM281) and cannabinoid 2 receptor (AM630) antagonists. CP55940 also caused several behavioral side effects, and these were prevented by the cannabinoid 1 receptor but not by the cannabinoid 2 receptor antagonist. Intrathecal injection of the cannabinoid 2 receptor agonist JWH015 reversed postoperative hypersensitivity (89 +/- 5% maximum possible effect; P < 0.05), and this was reversed by the cannabinoid 2 but not by the cannabinoid 1 receptor antagonist. JWH015, which did not induce behavioral side effects, reduced paw incision induced microglial and astrocytic activation in spinal cord (P < 0.05). CONCLUSIONS: These data indicate that intrathecal administration of cannabinoid receptor agonists may provide postoperative analgesia while reducing spinal glial activation, and that selective cannabinoid 2 receptor agonists may do so without central side effects.


Subject(s)
Pain, Postoperative/drug therapy , Receptor, Cannabinoid, CB2/agonists , Spinal Cord/physiology , Animals , Cyclohexanes/therapeutic use , Cyclohexanols , Indoles/therapeutic use , Male , Neuroglia/drug effects , Neuroglia/physiology , Phenols/therapeutic use , Rats , Rats, Sprague-Dawley , Reflex/drug effects
11.
Neuroreport ; 18(1): 67-71, 2007 Jan 08.
Article in English | MEDLINE | ID: mdl-17259863

ABSTRACT

Alpha 2-adrenoceptors are concentrated near sites of peripheral nerve injury or inflammation, primarily on immune cells, and their activation reduces inflammation and hypersensitivity to tactile stimuli. These results were obtained during acute inflammation, but the efficacy of alpha2-adrenoceptor stimulation in persistent inflammation has not been tested. Here, we show that perineural injection of the alpha2-adrenoceptor agonist, clonidine, reduces hypersensitivity in persistent sciatic neuritis with an onset more rapid than acute neuritis. Perineural clonidine reduces microglial activation in the spinal cord in persistent, but not acute neuritis, and does not change the number of spinal neurons with phosphorylated transcription factor, cyclic adenosine monophosphate response element binding protein. These data support treatment strategies with alpha2-adrenoceptor agonists in persistent neuritis.


Subject(s)
Analgesics/administration & dosage , Clonidine/administration & dosage , Sciatic Neuropathy/drug therapy , Animals , Behavior, Animal , Calcium-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Male , Microfilament Proteins , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Wistar , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology , Zymosan/administration & dosage
12.
Brain Behav Immun ; 21(5): 569-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17079113

ABSTRACT

Perineural alpha2-adrenoceptor activation relieves hypersensitivity induced by peripheral nerve injury or sciatic inflammatory neuritis. This effect is associated with a reduction in pro-inflammatory cytokines, as well as a reduction in local leukocyte number and their capacity to produce pro-inflammatory cytokines. Curiously, clonidine's antinociceptive effect appears with a 2-3-day delay after injection. Previous observations have shown that alpha-adrenoceptor activation induces apoptosis in leukocytes, which would reduce leukocyte number. Additionally, macrophage scavenging of apoptotic cells results in a shift to an anti-inflammatory phenotype, with expression of transforming growth factor (TGF)-beta1. We therefore examined the effects of perineural clonidine 24 h and 3 days after its injection on apoptosis, TGF-beta1 expression and lymphocyte and macrophage phenotype in acute sciatic inflammatory neuritis. Perineural clonidine reduced ipsilateral neuritis-induced hypersensitivity in a delayed manner (3 days after treatment), along with a reduction at this time in lymphocyte number and an increase in caspase-3 and TGF-beta1 expressing cells and macrophages co-expressing TGF-beta1 in the sciatic nerve. One day after injection clonidine treatment was associated with a reduction in lymphocytes and pro-inflammatory Th-1 cells as well as increased numbers of caspase-3 and TGF-beta1 expressing cells and macrophages co-expressing TGF-beta1 in sciatic nerve. Clonidine's effects were prevented by co-administration of an alpha2-adrenoceptor antagonist. These data suggest that alpha2-adrenoceptor activation in sciatic inflammatory neuritis increases local apoptosis and anti-inflammatory products early after treatment. This early effect likely underlies the delayed anti-inflammatory and anti-hypersensitivity effects of perineural clonidine in this setting.


Subject(s)
Analgesics/pharmacology , Apoptosis/immunology , Clonidine/pharmacology , Pain Threshold/drug effects , Sciatic Neuropathy/immunology , Adrenergic alpha-Agonists/immunology , Adrenergic alpha-Agonists/pharmacology , Analysis of Variance , Anesthesia, Local , Animals , Apoptosis/drug effects , Caspase 3/immunology , Caspase 3/metabolism , Cell Movement/drug effects , Clonidine/immunology , Disease Models, Animal , Leukocytes/drug effects , Leukocytes/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Pain Threshold/physiology , Rats , Rats, Wistar , Sciatic Nerve/cytology , Sciatic Nerve/drug effects , Sciatic Nerve/immunology , Sciatic Neuropathy/prevention & control , Statistics, Nonparametric , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/immunology
13.
Anesthesiology ; 104(2): 351-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436856

ABSTRACT

BACKGROUND: Partial sciatic nerve ligation (PSNL) produces axonal damage, a local inflammatory response, and wallerian degeneration. Cytokines secreted near the site of nerve injury are thought to play important roles in development and maintenance of central sensitization and neuropathic pain. Injection of clonidine at the site and time of nerve injury slows the development of PSNL-induced hypersensitivity and reduces local cytokine expression by actions on alpha2 adrenoceptors. The current study tested whether clonidine would have a similar effect in established nerve injury. METHODS: Rats underwent unilateral PSNL, and perineural saline, clonidine, or BRL44408 plus clonidine was injected 2 weeks later. Three days after perineural injection, withdrawal threshold to mechanical stimulation of the hind paw ipsilateral and contralateral to PSNL was determined, and tissues were removed for cytokine analysis. RESULTS: PSNL was accompanied by a proinflammatory pattern of cytokine content in neural structures and hypersensitivity ipsilaterally with few changes contralaterally. Perineural clonidine, but not saline, partially reversed the hypersensitivity, accompanied by reduced concentrations of interleukin 6 and interleukin 1beta in the sciatic nerve. The effect of clonidine on hypersensitivity and these cytokines was blocked by the alpha2-adrenoceptor antagonist, BRL44408. CONCLUSIONS: These data suggest that perineural clonidine acts on alpha2 adrenoceptors to reduce hypersensitivity in established nerve injury, likely by an immunomodulatory mechanism, and may be effective in patients in the weeks after nerve injury.


Subject(s)
Adrenergic alpha-Agonists/therapeutic use , Clonidine/therapeutic use , Cytokines/biosynthesis , Pain/drug therapy , Sciatic Nerve/injuries , Sciatic Neuropathy/complications , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Antagonists/pharmacology , Animals , Behavior, Animal/drug effects , Clonidine/administration & dosage , Imidazoles/pharmacology , Indoles/pharmacology , Injections , Isoindoles , Ligation , Male , Pain/etiology , Pain/psychology , Pain Threshold/drug effects , Physical Stimulation , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...