1
|
Drinovac Vlah V, Bach-Rojecky L. Mirror-Image Pain Update: Complex Interactions Between Central and Peripheral Mechanisms. Mol Neurobiol 2024; 61:1-18. [PMID: 38602655 DOI: 10.1007/s12035-024-04102-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
The appearance of contralateral effects after unilateral injury has been shown in various experimental pain models, as well as in clinics. They consist of a diversity of phenomena in contralateral peripheral nerves, sensory ganglia, or spinal cord: from structural changes and altered gene or protein expression to functional consequences such as the development of mirror-image pain (MP). Although MP is a well-documented phenomenon, the exact molecular mechanism underlying the induction and maintenance of mirror-like spread of pain is still an unresolved challenge. MP has generally been explained by central sensitization mechanisms leading to facilitation of pain impulse transfer through neural connections between the two sides of the central nervous system. On the contrary, the peripheral nervous system (PNS) was usually regarded unlikely to evoke such a symmetrical phenomenon. However, recent findings provided evidence that events in the PNS could play a significant role in MP induction. This manuscript provides an updated and comprehensive synthesis of the MP phenomenon and summarizes the available data on the mechanisms. A more detailed focus is placed on reported evidence for peripheral mechanisms behind the MP phenomenon, which were not reviewed up to now.
Collapse
Affiliation(s)
- Višnja Drinovac Vlah
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, Domagojeva 2, 10000, Zagreb, Croatia
| | - Lidija Bach-Rojecky
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, Domagojeva 2, 10000, Zagreb, Croatia.
| |
Collapse
|
2
|
Casaril AM, Gaffney CM, Shepherd AJ. Animal models of neuropathic pain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:339-401. [PMID: 39580217 DOI: 10.1016/bs.irn.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Animal models continue to be crucial to developing our understanding of the molecular, cellular, and neurophysiological mechanisms that lead to neuropathic pain. The overwhelming majority of animal studies use rodent models, ranging from surgical and trauma-induced models to those induced by metabolic diseases, genetic mutations, viruses, neurotoxic drugs, and cancer. We discuss the clinical relevance of the available models and the pain behavior tests commonly used as outcome measures. Finally, we summarize the refinements that have been proposed to improve the ability of animal model studies to predict clinical efficacy.
Collapse
Affiliation(s)
- Angela M Casaril
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Caitlyn M Gaffney
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrew J Shepherd
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
3
|
Vincenzi M, Milella MS, D’Ottavio G, Caprioli D, Reverte I, Maftei D. Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain. Life (Basel) 2022; 12:life12030398. [PMID: 35330149 PMCID: PMC8955776 DOI: 10.3390/life12030398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Neuropathic pain (NP) originates from an injury or disease of the somatosensory nervous system. This heterogeneous origin and the possible association with other pathologies make the management of NP a real challenge. To date, there are no satisfactory treatments for this type of chronic pain. Even strong opioids, the gold-standard analgesics for nociceptive and cancer pain, display low efficacy and the paradoxical ability to exacerbate pain sensitivity in NP patients. Mounting evidence suggests that chemokine upregulation may be a common mechanism driving NP pathophysiology and chronic opioid use-related consequences (analgesic tolerance and hyperalgesia). Here, we first review preclinical studies on the role of chemokines and chemokine receptors in the development and maintenance of NP. Second, we examine the change in chemokine expression following chronic opioid use and the crosstalk between chemokine and opioid receptors. Then, we examine the effects of inhibiting specific chemokines or chemokine receptors as a strategy to increase opioid efficacy in NP. We conclude that strong opioids, along with drugs that block specific chemokine/chemokine receptor axis, might be the right compromise for a favorable risk/benefit ratio in NP management.
Collapse
Affiliation(s)
- Martina Vincenzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (M.V.); (I.R.)
| | - Michele Stanislaw Milella
- Toxicology and Poison Control Center Unit, Department of Emergency, Anesthesia and Critical Care, Policlinico Umberto I Hospital-Sapienza University of Rome, 00161 Rome, Italy;
| | - Ginevra D’Ottavio
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Daniele Caprioli
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Ingrid Reverte
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Correspondence: (M.V.); (I.R.)
| | - Daniela Maftei
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
| |
Collapse
|
4
|
Eidson LN, Murphy AZ. Inflammatory mediators of opioid tolerance: Implications for dependency and addiction. Peptides 2019; 115:51-58. [PMID: 30890355 PMCID: PMC6863079 DOI: 10.1016/j.peptides.2019.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/27/2022]
Abstract
Each year, over 50 million Americans suffer from persistent pain, including debilitating headaches, joint pain, and severe back pain. Although morphine is amongst the most effective analgesics available for the management of severe pain, prolonged morphine treatment results in decreased analgesic efficacy (i.e., tolerance). Despite significant headway in the field, the mechanisms underlying the development of morphine tolerance are not well understood. The midbrain ventrolateral periaqueductal gray (vlPAG) is a primary neural substrate for the analgesic effects of morphine, as well as for the development of morphine tolerance. A growing body of literature indicates that activated glia (i.e., microglia and astrocytes) facilitate pain transmission and oppose morphine analgesia, making these cells important potential targets in the treatment of chronic pain. Morphine affects glia by binding to the innate immune receptor toll-like receptor 4 (TLR4), leading to the release of proinflammatory cytokines and opposition of morphine analgesia. Despite the established role of the vlPAG as an integral locus for the development of morphine tolerance, most studies have examined the role of glia activation within the spinal cord. Additionally, the role of TLR4 in the development of tolerance has not been elucidated. This review attempts to summarize what is known regarding the role of vlPAG glia and TLR4 in the development of morphine tolerance. These data, together, provide information about the mechanism by which central nervous system glia regulate morphine tolerance, and identify a potential therapeutic target for the enhancement of analgesic efficacy in the clinical treatment of chronic pain.
Collapse
Affiliation(s)
- Lori N Eidson
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30308, United States.
| |
Collapse
|
5
|
A single peri-sciatic nerve administration of the adenosine 2A receptor agonist ATL313 produces long-lasting anti-allodynia and anti-inflammatory effects in male rats. Brain Behav Immun 2019; 76:116-125. [PMID: 30453021 DOI: 10.1016/j.bbi.2018.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/09/2018] [Accepted: 11/13/2018] [Indexed: 01/08/2023] Open
Abstract
Neuropathic pain is a widespread problem which remains poorly managed by currently available therapeutics. Peripheral nerve injury and inflammation leads to changes at the nerve injury site, including activation of resident and recruited peripheral immune cells, that lead to neuronal central sensitization and pain amplification. The present series of studies tested the effects of peri-sciatic nerve delivery of single doses of adenosine 2A receptor (A2aR) agonists on pain and neuroinflammation. The data provide converging lines of evidence supportive that A2aR agonism at the site of peripheral nerve injury and inflammation is effective in suppressing ongoing neuropathic pain. After A2aR agonism resolved neuropathic pain, a return of pain enhancement (allodynia) was observed in response to peri-sciatic injection of H-89, which can inhibit protein kinase A, and by peri-sciatic injection of neutralizing antibody against the potent anti-inflammatory cytokine interleukin-10. A2aR agonist actions at the nerve injury site suppress neuroinflammation, as reflected by decreased release of interleukin-1β and nitric oxide, as well as decreased sciatic expression of markers of monocytes/macrophages and inducible nitric oxide synthase. Taken together, the data are supportive that A2aR agonists, acting at the level of peripheral nerve injury, may be of therapeutic value in treating chronic pain of neuroinflammatory origin.
Collapse
|
6
|
Young A, Kalladka M, Viswanath A, Zusman T, Khan J. Consomic rats parental strains differ in sensory perception, pain developed following nerve injury and in IL-1 beta and IL-6 levels. ACTA ACUST UNITED AC 2018; 25:137-141. [PMID: 29580677 DOI: 10.1016/j.pathophys.2018.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/12/2018] [Accepted: 03/18/2018] [Indexed: 11/26/2022]
Affiliation(s)
- Andrew Young
- Orofacial Disorders Clinic, Department of Diagnostic Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, 155 Fifth Street, San Francisco, CA, 94103, USA.
| | - Mythili Kalladka
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, 625 Elmwood Ave, Rochester, NY, 14620, USA.
| | - Archana Viswanath
- Department of Oral and Maxillofacial Surgery, Tufts University School of Dental Medicine, USA.
| | - Tal Zusman
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ, USA.
| | - Junad Khan
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, 625 Elmwood Ave, Rochester, NY, 14620, USA.
| |
Collapse
|
7
|
Kumar A, Kaur H, Singh A. Neuropathic Pain models caused by damage to central or peripheral nervous system. Pharmacol Rep 2017; 70:206-216. [PMID: 29475003 DOI: 10.1016/j.pharep.2017.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/24/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Neuropathic Pain (NP) is a painful condition which is a direct consequence of a lesion or disease affecting the somatosensory system with symptoms like allodynia, hyperalgesia. It has complex pathogenesis as it involves several molecular signaling pathways, thus numerous reliable animal models are crucial to understand the underlying mechanism of NP and formulate effective management therapy. Some models like spinal cord injury, chronic constriction injury, spinal nerve ligation, chemotherapy induced peripheral neuropathy, diabetes-induced NP and many more are discussed. This review contains an overview of the procedures followed to induce neuropathy and specific characteristics of that particular model. Some new techniques like spared nerve ligation, have omitted the limitation of methods not presently used where complete nerve damage occurs. Since animal models provide a window to experienced symptoms and physiology and impact the translation of bench discoveries to the bedside, the reporting, interpretation and comparison of these models is necessary because slight variation in procedure of model generation can drastically alter the results. The development of novel, but rational analgesic drugs to alleviate this intractable pain demands elucidation of molecular mechanisms of NP for which different types of animal models have been established.
Collapse
Affiliation(s)
- Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India.
| | - Harshpreet Kaur
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| | - Arti Singh
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| |
Collapse
|
8
|
Khan J, Hassun H, Zusman T, Korczeniewska O, Eliav E. Interleukin-8 levels in rat models of nerve damage and neuropathic pain. Neurosci Lett 2017; 657:106-112. [PMID: 28789985 DOI: 10.1016/j.neulet.2017.07.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/27/2017] [Indexed: 01/15/2023]
Abstract
Interleukin-8 (IL-8) is a pro-inflammatory cytokine that has been shown to play a role in inflammatory and autoimmune disorders. The objective of the present study was to assess the levels of IL-8 in rat serum, dorsal root ganglion (DRG) and the sciatic nerve following four different forms of sciatic nerve injury. The models used to induce the injury included partial sciatic ligation (PSL), chronic constriction injury (CCI), perineural inflammation (neuritis) and complete sciatic transection (CST). Mechanical and thermal hyperalgesia were detected by measuring withdrawal responses from a mechanical stimulus and withdrawal latency from thermal stimulation. Enzyme-linked immunosorbent assays (ELISA) was used to assess the IL-8 levels in the affected and contralateral sciatic nerves. Rats exposed to PSL and neuritis developed significant nociceptive response (mechanical and thermal hyperalgesia) in the affected side at three days post-surgery whereas the CCI group at eight days post-surgery. No mechanical or thermal hyperalgesia was observed in rats exposed to CST at either three or eight days postsurgery. Additionally, IL-8 levels were significantly increased in the injured sciatic nerve at 3 and 8days following PSL and neuritis as well as at 8days following CCI when compared to naïve animals. A significant up regulation of IL-8 levels was observed in the ipsilateral DRG at 3 and 8days following CST compared to naïve animals. The serum IL-8 levels remained unchanged in all models of nerve damage. The results of this study suggest that IL-8's role in the neuropathic pain etiology may be specific to nerve injury type.
Collapse
Affiliation(s)
- Junad Khan
- Eastman Institute for Oral Health, Rochester, NY, 14620, United States.
| | - Humza Hassun
- Department of Diagnostic Sciences, Rutgers School of Dental Medicine, NJ, United States
| | - Tali Zusman
- Department of Diagnostic Sciences, Rutgers School of Dental Medicine, NJ, United States
| | - Olga Korczeniewska
- Department of Diagnostic Sciences, Rutgers School of Dental Medicine, NJ, United States
| | - Eli Eliav
- Eastman Institute for Oral Health, University of Rochester, NY, United States
| |
Collapse
|
9
|
McIntyre MK, Clifford JL, Maani CV, Burmeister DM. Progress of clinical practice on the management of burn-associated pain: Lessons from animal models. Burns 2016; 42:1161-72. [PMID: 26906668 DOI: 10.1016/j.burns.2016.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/05/2016] [Accepted: 01/21/2016] [Indexed: 02/08/2023]
Abstract
Opioid-based analgesics provide the mainstay for attenuating burn pain, but they have a myriad of side effects including respiratory depression, nausea, impaired gastrointestinal motility, sedation, dependence, physiologic tolerance, and opioid-induced hyperalgesia. To test and develop novel analgesics, validated burn-relevant animal models of pain are indispensable. Herein we review such animal models, which are mostly limited to rodent models of burn-induced, inflammatory, and neuropathic pain. The latter two are pain syndromes that provide insight into the pain caused by systemic pro-inflammatory cytokines and direct injury to nerves (e.g., after severe burn), respectively. To date, no single animal model optimally mimics the complex pathophysiology and pain that a human burn patient experiences. No currently available burn-pain model examines effects of pharmacological intervention on wound healing. As cornerstones of pain and wound healing, pro-inflammatory mediators may be utilized for insight into both processes. Moreover, common clinical concerns such as systemic inflammatory response syndrome and multiple organ dysfunction remain unaddressed. For development of analgesics, these aberrations can significantly alter the potential efficacy and/or adverse effects of a prescribed analgesic following burn trauma. We therefore suggest that a multi-model strategy would be the most clinically relevant when evaluating novel analgesics for use in burn patients.
Collapse
Affiliation(s)
- Matthew K McIntyre
- United States Army Institute of Surgical Research, 3650 Chambers Pass, BHT1:Bldg 3610, JBSA Fort Sam Houston, TX 78234-6315, United States.
| | - John L Clifford
- United States Army Institute of Surgical Research, 3650 Chambers Pass, BHT1:Bldg 3610, JBSA Fort Sam Houston, TX 78234-6315, United States.
| | - Christopher V Maani
- Brooke Army Medical Center, Department of Anesthesia and Operative Services, San Antonio Uniformed Services Health Education Consortium (SAUSHEC) Anesthesia Residency Program, 3551 Roger Brooke Drive, JBSA Fort Sam Houston, TX 78234-6315, United States.
| | - David M Burmeister
- United States Army Institute of Surgical Research, 3650 Chambers Pass, BHT1:Bldg 3610, JBSA Fort Sam Houston, TX 78234-6315, United States.
| |
Collapse
|
10
|
Ceusters W, Nasri-Heir C, Alnaas D, Cairns BE, Michelotti A, Ohrbach R. Perspectives on next steps in classification of oro-facial pain - Part 3: biomarkers of chronic oro-facial pain - from research to clinic. J Oral Rehabil 2015; 42:956-66. [PMID: 26200973 PMCID: PMC4715524 DOI: 10.1111/joor.12324] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2015] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to review the current status of biomarkers used in oro-facial pain conditions. Specifically, we critically appraise their relative strengths and weaknesses for assessing mechanisms associated with the oro-facial pain conditions and interpret that information in the light of their current value for use in diagnosis. In the third section, we explore biomarkers through the perspective of ontological realism. We discuss ontological problems of biomarkers as currently widely conceptualised and implemented. This leads to recommendations for research practice aimed to a better understanding of the potential contribution that biomarkers might make to oro-facial pain diagnosis and thereby fulfil our goal for an expanded multidimensional framework for oro-facial pain conditions that would include a third axis.
Collapse
Affiliation(s)
- Werner Ceusters
- Department of Biomedical Informatics, University at Buffalo, NY, USA
| | | | | | - Brian E Cairns
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Ambra Michelotti
- Section of Orthodontics, School of Dentistry, University of Naples Federico II, Naples, Italy
| | - Richard Ohrbach
- Department of Oral Diagnostic Sciences, University at Buffalo, NY, USA
| |
Collapse
|
11
|
Khan J, Ramadan K, Korczeniewska O, Anwer MM, Benoliel R, Eliav E. Interleukin-10 levels in rat models of nerve damage and neuropathic pain. Neurosci Lett 2015; 592:99-106. [DOI: 10.1016/j.neulet.2015.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/25/2015] [Accepted: 03/01/2015] [Indexed: 12/30/2022]
|
12
|
Page GG, Opp MR, Kozachik SL. Reduced sleep, stress responsivity, and female sex contribute to persistent inflammation-induced mechanical hypersensitivity in rats. Brain Behav Immun 2014; 40:244-51. [PMID: 24594386 DOI: 10.1016/j.bbi.2014.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 01/22/2023] Open
Abstract
Studies in humans suggest that female sex, reduced sleep opportunities and biological stress responsivity increase risk for developing persistent pain conditions. To investigate the relative contribution of these three factors to persistent pain, we employed the Sciatic Inflammatory Neuritis (SIN) model of repeated left sciatic perineurial exposures to zymosan, an inflammatory stimulus, to determine their impact upon the development of persistent mechanical hypersensitivity. Following an initial moderate insult, a very low zymosan dose was infused daily for eight days to model a sub-threshold inflammatory perturbation to which only susceptible animals would manifest or maintain mechanical hypersensitivity. Using Sprague Dawley rats, maintaining wakefulness throughout the first one-half of the 12-h light phase resulted in a bilateral reduction in paw withdrawal thresholds (PWTs); zymosan infusion reduced ipsilateral PWTs in all animals and contralateral PWTs only in females. This sex difference was validated in Fischer 344, Lewis and Sprague Dawley rats, suggesting that females are the more susceptible phenotype for both local and centrally driven responses to repeated low-level inflammatory perturbations. Hypothalamic-pituitary-adrenal (HPA) axis hyporesponsive Lewis rats exhibited the most robust development of mechanical hypersensitivity and HPA axis hyperresponsive Fischer 344 rats matched the Lewis rats' mechanical hypersensitivity throughout the latter four days of the protocol. If HPA axis phenotype does indeed influence these findings, the more balanced responsivity of Sprague Dawley rats would seem to promote resilience in this paradigm. Taken together, these findings are consistent with what is known regarding persistent pain development in humans.
Collapse
Affiliation(s)
- Gayle G Page
- School of Nursing, Johns Hopkins University, 525 N. Wolfe St., Baltimore, MD 21205, United States.
| | - Mark R Opp
- Department of Anesthesiology & Pain Medicine, University of Washington, 325 9th Ave, Box #359724, Seattle, WA 98104, United States.
| | - Sharon L Kozachik
- School of Nursing, Johns Hopkins University, 525 N. Wolfe St., Baltimore, MD 21205, United States.
| |
Collapse
|
13
|
Blood-nerve barrier dysfunction contributes to the generation of neuropathic pain and allows targeting of injured nerves for pain relief. Pain 2014; 155:954-967. [DOI: 10.1016/j.pain.2014.01.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/15/2014] [Accepted: 01/28/2014] [Indexed: 01/09/2023]
|
14
|
Gear RW, Bogen O, Ferrari LF, Green PG, Levine JD. NOP receptor mediates anti-analgesia induced by agonist-antagonist opioids. Neuroscience 2014; 257:139-48. [PMID: 24188792 PMCID: PMC3947912 DOI: 10.1016/j.neuroscience.2013.10.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 10/25/2013] [Accepted: 10/25/2013] [Indexed: 11/18/2022]
Abstract
Clinical studies have shown that agonist-antagonist opioid analgesics that produce their analgesic effect via action on the kappa-opioid receptor, produce a delayed-onset anti-analgesia in men but not women, an effect blocked by co-administration of a low dose of naloxone. We now report the same time-dependent anti-analgesia and its underlying mechanism in an animal model. Using the Randall-Selitto paw-withdrawal assay in male rats, we found that nalbuphine, pentazocine, and butorphanol each produced analgesia during the first hour followed by anti-analgesia starting at ∼90min after administration in males but not females, closely mimicking its clinical effects. As observed in humans, co-administration of nalbuphine with naloxone in a dose ratio of 12.5:1 blocked anti-analgesia but not analgesia. Administration of the highly selective kappa-opioid receptor agonist U69593 produced analgesia without subsequent anti-analgesia, and confirmed by the failure of the selective kappa antagonist nor-binaltorphimine to block nalbuphine-induced anti-analgesia, indicating that anti-analgesia is not mediated by kappa-opioid receptors. We therefore tested the role of other receptors in nalbuphine anti-analgesia. Nociceptin/orphanin FQ (NOP) and sigma-1 and sigma-2 receptors were chosen on the basis of their known anti-analgesic effects and receptor binding studies. The selective NOP receptor antagonists, JTC801, and J-113397, but not the sigma receptor antagonist, BD 1047, antagonized nalbuphine anti-analgesia. Furthermore, the NOP receptor agonist NNC 63-0532 produced anti-analgesia with the same delay in onset observed with the three agonist-antagonists, but without producing preceding analgesia and this anti-analgesia was also blocked by naloxone. These results strongly support the suggestion that clinically used agonist-antagonists act at the NOP receptor to produce anti-analgesia.
Collapse
Affiliation(s)
- R W Gear
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, CA 94143-0440, United States
| | - O Bogen
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, CA 94143-0440, United States
| | - L F Ferrari
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, CA 94143-0440, United States
| | - P G Green
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, CA 94143-0440, United States
| | - J D Levine
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, CA 94143-0440, United States; Department of Medicine, University of California at San Francisco, San Francisco, CA 94143-0120, United States.
| |
Collapse
|
15
|
Abstract
Chronic pain affects as many as one in five people. A proportion of patients with symptoms of neuropathic -pain do not have clinical signs of any obvious tissue or nerve injury. Such patients include those with diffuse limb pain, back pain, and complex regional pain syndrome type 1. These patients remain a clinical enigma. However, through the development of the neuritis model, it has become apparent that local nerve inflammation in the absence of gross pathology (i.e., axonal degeneration and demyelination) may underlie part of the mechanisms of pain. In this chapter, we describe a method to induce the neuritis model. We also describe in detail a reliable method to test for mechanical allodynia and heat hyperalgesia. Data that demonstrates the potential benefits of the neuroprotective agent ARA290 in reducing pain behavior in the neuritis model are presented.
Collapse
|
16
|
Pitcher GM, Ritchie J, Henry JL. Peripheral neuropathy induces cutaneous hypersensitivity in chronically spinalized rats. PAIN MEDICINE 2013; 14:1057-71. [PMID: 23855791 DOI: 10.1111/pme.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES The present study was aimed at the issue of whether peripheral nerve injury-induced chronic pain is maintained by supraspinal structures governing descending facilitation to the spinal dorsal horn, or whether altered peripheral nociceptive mechanisms sustain central hyperexcitability and, in turn, neuropathic pain. We examined this question by determining the contribution of peripheral/spinal mechanisms, isolated from supraspinal influence(s), in cutaneous hypersensitivity in an animal model of peripheral neuropathy. METHODS Adult rats were spinalized at T8-T9; 8 days later, peripheral neuropathy was induced by implanting a 2-mm polyethylene cuff around the left sciatic nerve. Hind paw withdrawal responses to mechanical or thermal plantar stimulation were evaluated using von Frey filaments or a heat lamp, respectively. RESULTS Spinalized rats without cuff implantation exhibited a moderate decrease in mechanical withdrawal threshold on ~day 10 (P < 0.05) and in thermal withdrawal threshold on ~day 18 (P < 0.05). However, cuff-implanted spinalized rats developed a more rapid and significant decrease in mechanical (~day 4; P < 0.001) and thermal (~day 10; P < 0.05) withdrawal thresholds that remained significantly decreased through the duration of the study. CONCLUSIONS Our findings demonstrate an aberrant peripheral/spinal mechanism that induces and maintains thermal and to a greater degree tactile cutaneous hypersensitivity in the cuff model of neuropathic pain, and raise the prospect that altered peripheral/spinal nociceptive mechanisms in humans with peripheral neuropathy may have a pathologically relevant role in both inducing and sustaining neuropathic pain.
Collapse
Affiliation(s)
- Graham M Pitcher
- Departments of Physiology and Psychiatry, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
17
|
Nie F, Wang J, Su D, Shi Y, Chen J, Wang H, Qin W, Shi L. Abnormal activation of complement C3 in the spinal dorsal horn is closely associated with progression of neuropathic pain. Int J Mol Med 2013; 31:1333-42. [PMID: 23588254 DOI: 10.3892/ijmm.2013.1344] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 03/20/2013] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the role of complement activation in the pathogenesis of neuropathic pain (NPP) induced by peripheral nerve injury. We modified a classical chronic constriction injury (CCI) model (mCCI), and verified its reliability in rats. Furthermore, reverse transcription-PCR and immunohistochemistry were conducted to investigate complement activation in the spinal dorsal horn and the effect of a complement inhibitor, cobra venom factor (CVF), on the behavior of the mCCI model rats. We found that rats in the mCCI group presented a better general condition, without signs of autophagy of the toes. Moreover, mCCI induced a significant increase (+40%) in the expression of component 3 (C3) mRNA in the spinal dorsal horn, which was associated with hyperalgesia. Correlation analysis showed a negative correlation between the mechanical pain threshold and the expression of C3 in the spinal cord. Administration of CVF reduced the occurrence of hyperalgesia in mCCI rats and nearly reversed the hyperalgesia. In addition, the mCCI rats exhibited significantly less spinal superoxide dismutase activity and significantly greater levels of maleic dialdehyde compared to the sham-operated rats. Transmission electron micrographs revealed mitochondrial swelling, cell membrane damage, and cristae fragmentation in the neurons of the spinal dorsal horn 14 days after mCCI. Mitochondrial swelling was attenuated in mCCI rats receiving CVF. The findings demonstrated that abnormal complement activation occurred in the dorsal horn of the spinal cord in rats with NPP, and C3 in the spinal dorsal horn could play an important role in the cascade reaction of complements that are involved in the development of hyperalgesia.
Collapse
Affiliation(s)
- Fachuan Nie
- Department of Pain Care and Nonvascular Intervention, Third Military Medical University, Chongqing, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Eidson LN, Murphy AZ. Persistent peripheral inflammation attenuates morphine-induced periaqueductal gray glial cell activation and analgesic tolerance in the male rat. THE JOURNAL OF PAIN 2013; 14:393-404. [PMID: 23395474 PMCID: PMC3991566 DOI: 10.1016/j.jpain.2012.12.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/12/2012] [Accepted: 12/14/2012] [Indexed: 11/15/2022]
Abstract
UNLABELLED Morphine is among the most prevalent analgesics prescribed for chronic pain. However, prolonged morphine treatment results in the development of analgesic tolerance. An abundance of evidence has accumulated indicating that central nervous system glial cell activity facilitates pain transmission and opposes morphine analgesia. While the midbrain ventrolateral periaqueductal gray (vlPAG) is an important neural substrate mediating pain modulation and the development of morphine tolerance, no studies have directly assessed the role of PAG glia. Here we test the hypothesis that morphine-induced increases in vlPAG glial cell activity contribute to the development of morphine tolerance. As morphine is primarily consumed for the alleviation of severe pain, the influence of persistent inflammatory pain was also assessed. Administration of morphine, in the absence of persistent inflammatory pain, resulted in the rapid development of morphine tolerance and was accompanied by a significant increase in vlPAG glial activation. In contrast, persistent inflammatory hyperalgesia, induced by intraplantar administration of complete Freund's adjuvant (CFA), significantly attenuated the development of morphine tolerance. No significant differences were noted in vlPAG glial cell activation for CFA-treated animals versus controls. These results indicate that vlPAG glia are modulated by a persistent pain state, and implicate vlPAG glial cells as possible regulators of morphine tolerance. PERSPECTIVE The development of morphine tolerance represents a significant impediment to its use in the management of chronic pain. We report that morphine tolerance is accompanied by increased glial cell activation within the vlPAG, and that the presence of a persistent pain state prevented vlPAG glial activation and attenuated morphine tolerance.
Collapse
Affiliation(s)
- Lori N Eidson
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303, USA
| | | |
Collapse
|
19
|
|
20
|
Richards N, Batty T, Dilley A. CCL2 has similar excitatory effects to TNF-α in a subgroup of inflamed C-fiber axons. J Neurophysiol 2011; 106:2838-48. [PMID: 21865436 PMCID: PMC3234089 DOI: 10.1152/jn.00183.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peripheral nerve inflammation can cause neuronal excitability changes that have been implicated in the pathogenesis of chronic pain. Although the neuroimmune interactions that lead to such physiological changes are unclear, in vitro studies suggest that the chemokine CCL2 may be involved. This in vivo study examines the effects of CCL2 on untreated and inflamed neurons and compares its effects with those of TNF-α. Extracellular recordings were performed in the anesthetized rat on isolated neurons with C-fiber axons. On untreated neurons, CCL2, as well as TNF-α, had negligible effects. Following neuritis, both cytokines transiently caused the firing of action potentials in 27–30% of neurons, which were either silent or had background (ongoing) activity. The neurons with ongoing activity, which responded to either cytokine, had significantly slower baseline firing rates {median = 3.0 spikes/min [interquartile range (IQR) 3.0]} compared with the nonresponders [median = 24.4 spikes/min (IQR 24.6); P < 0.001]. In an additional group, 26–27% of neurons, which were sensitized due to repeated noxious mechanical stimulation of the periphery, also responded to the effects of both cytokines. Neither cytokine caused axons to become mechanically sensitive. Immunohistochemistry confirmed that the cognate CCL2 receptor, CCR2, is mainly expressed on glia and is therefore not likely to be an axonal target for CCL2 following inflammation. In contrast, the cognate TNF-α receptor (TNFR), TNFR1, was present on untreated and inflamed neurons. In summary, CCL2 can excite inflamed C-fiber neurons with similar effects to TNF-α, although the underlying mechanisms may be different. The modulatory effects of both cytokines are limited to a subgroup of neurons, which may be subtly inflamed.
Collapse
Affiliation(s)
- Natalie Richards
- Division of Clinical and Laboratory Investigation, Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, UK
| | | | | |
Collapse
|
21
|
|
22
|
Experimental model of zymosan-induced arthritis in the rat temporomandibular joint: role of nitric oxide and neutrophils. J Biomed Biotechnol 2011; 2011:707985. [PMID: 21274271 PMCID: PMC3022228 DOI: 10.1155/2011/707985] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 12/02/2010] [Accepted: 12/09/2010] [Indexed: 11/17/2022] Open
Abstract
AIMS To establish a new model of zymosan-induced temporomandibular joint (TMJ) arthritis in the rat and to investigate the role of nitric oxide. METHODS Inflammation was induced by an intra-articular injection of zymosan into the left TMJ. Mechanical hypernociception, cell influx, vascular permeability, myeloperoxidase activity, nitrite levels, and histological changes were measured in TMJ lavages or tissues at selected time points. These parameters were also evaluated after treatment with the nitric oxide synthase (NOS) inhibitors L-NAME or 1400 W. RESULTS Zymosan-induced TMJ arthritis caused a time-dependent leucocyte migration, plasma extravasation, mechanical hypernociception, and neutrophil accumulation between 4 and 24 h. TMJ immunohistochemical analyses showed increased inducible NOS expression. Treatment with L-NAME or 1400 W inhibited these parameters. CONCLUSION Zymosan-induced TMJ arthritis is a reproducible model that may be used to assess both the mechanisms underlying TMJ inflammation and the potential tools for therapies. Nitric oxide may participate in the inflammatory temporomandibular dysfunction mechanisms.
Collapse
|
23
|
Elliott M, Barr A, Clark B, Wade C, Barbe M. Performance of a repetitive task by aged rats leads to median neuropathy and spinal cord inflammation with associated sensorimotor declines. Neuroscience 2010; 170:929-41. [DOI: 10.1016/j.neuroscience.2010.07.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 07/21/2010] [Accepted: 07/21/2010] [Indexed: 12/30/2022]
|
24
|
Eliav E, Benoliel R, Herzberg U, Kalladka M, Tal M. The role of IL-6 and IL-1beta in painful perineural inflammatory neuritis. Brain Behav Immun 2009; 23:474-84. [PMID: 19486649 DOI: 10.1016/j.bbi.2009.01.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 01/05/2009] [Accepted: 01/15/2009] [Indexed: 10/21/2022] Open
Abstract
UNLABELLED Inflammation along a nerve trunk (perineural inflammation), without detectable axonal damage, has been shown to induce transient pain in the organ supplied by the nerve. The aims of the present study were to study the role IL-6 and IL-1beta, in pain induced by perineural inflammation. METHODS IL-6 and IL-1beta secretion from rat's sciatic nerves, L-5 Dorsal Root Ganglia (DRG), and the hind paw skin, 3 and 8 days following exposure of the nerve to Complete Freund's Adjuvant (CFA), were measured using ELISA method. Hind paw tactile-allodynia, mechano-hyperalgesia, heat-allodynia and electrical detection thresholds were tested up to 8 days following the application of CFA, IL-6 or IL-1beta adjacent to the sciatic nerve trunk. Employing electrophysiological recording, saphenous nerve spontaneous activity, nerve trunk mechano-sensitivity and paw tactile detection threshold (determined by recording action potential induced by the lowest mechanical stimulus) were assessed 3 and 8 days following exposure of the nerve trunk to CFA, IL-6, or IL-1beta. RESULTS IL-6 and IL-1beta secretion from the nerve was significantly elevated on the 3rd day post-operation (DPO). On the 8th DPO, IL-6 levels returned to baseline while IL-1beta levels remained significantly elevated. The DRG cytokine's level was increased on the 3rd and 8th DPOs, contralateral cytokine's level was increased on the 3rd DPO. The skin IL-6 level was increased bilaterally on the 3rd DPO and returned to baseline on the 8th DPO. IL-1beta levels increased in the affected side on the 3rd and bilaterally on the 8th DPO. Direct application of IL-6 or CFA on the sciatic nerve induced significant hind paw tactile-allodynia from the 1st to 5th DPOs, reduced electrical detection threshold from the 1st to 3rd DPOs, mechano-hyperalgesia from 3rd to 5th DPOs and heat-allodynia on the 3rd DPO. Direct application of IL-1beta induced paw tactile and heat-allodynia on the 7-8th DPOs and mechano-hyperalgesia on the 5-8th DPOs. Perineural inflammation significantly increased spontaneous activity myelinated fibres 3 and 8 days following the application. Direct application of IL-6 induced elevation of spontaneous activity on the 3rd while IL-1beta on the 8th DPO. Nerve mechano-sensitivity was significantly increased on the 3rd day following exposure to CFA and IL-6 and on the 8th following CFA application. The rat's paw lowest mechanical force necessary for induction of action potential, was significantly reduced 3 days following CFA application. CONCLUSION IL-6 and IL-1beta play an important role in pain induced by perineural inflammation. IL-6 activity is more prominent immediately following application (2-5th DPOs), while IL-1beta, activity is more significant in a later stage (5-8th DPOs).
Collapse
Affiliation(s)
- Eli Eliav
- UMDNJ-New Jersey Dental School, Carmel Endowed Chair in Algesiology, Department of Diagnostic Sciences, Division of Orofacial Pain, 110 Bergen Street, Newark, NJ 07103, USA.
| | | | | | | | | |
Collapse
|
25
|
Neural fractalkine expression is closely linked to pain and pancreatic neuritis in human chronic pancreatitis. J Transl Med 2009; 89:347-61. [PMID: 19153557 DOI: 10.1038/labinvest.2008.170] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The chemokine fractalkine induces migration of inflammatory cells into inflamed tissues, thereby aggravating inflammatory tissue damage and fibrosis. Furthermore, fractalkine increases neuropathic pain through glial activation, which can be diminished by blocking of its receptor, CX3CR1, through neutralizing antibodies. As chronic pancreatitis (CP) is characterized by tissue infiltration of inflammatory cells, fibrosis, pancreatic neuritis and severe pain, the roles of fractalkine and CX3CR1 were investigated in CP (n=61) and normal pancreas (NP, n=21) by QRT-PCR, western blot and immunohistochemistry analyses. Their expression correlated with the severity of pancreatic neuritis, fibrosis, intrapancreatic nerve fiber density and hypertrophy, pain, CP duration and with the amount of inflammatory cell infiltrate immuno-positive for CD45 and CD68. To investigate the influence of fractalkine on pancreatic fibrogenesis, human pancreatic stellate cells (hPSCs) were isolated from patients with CP, incubated with fractalkine and then Collagen-1 and alpha-smooth muscle actin (alpha-SMA) expressions were measured. CX3CR1, but not fractalkine, mRNA was overexpressed in CP. In contrast, the protein levels of both CX3CR1 and fractalkine were upregulated. Neuro-immunoreactivity for fractalkine and CX3CR1 was strongest in patients suffering from severe pain and pancreatic neuritis. Long-term suffering from CP was noticeably related to increased neural immunoreactivity of fractalkine. Furthermore, fractalkine and CX3CR1 mRNA overexpressions were associated with enhanced lymphocyte and macrophage infiltration. Advanced fibrosis was associated with increased fractalkine expression, whereas in vitro fractalkine had no significant impact on collagen-1 and alpha-SMA expressions in hPSCs. Therefore, pancreatic fractalkine expression appears to be linked to visceral pain and to the recruitment of inflammatory cells into the pancreatic tissue and nerve fibers, with subsequent pancreatic neuritis. However, pancreatic fibrogenesis is probably indirectly influenced by fractalkine. Taken together, these novel findings suggest that CX3CR1 represents a potential novel therapeutic target to reduce inflammation and modulate pain in CP.
Collapse
|
26
|
Models in Pain Research. Mol Pain 2009. [DOI: 10.1007/978-0-387-75269-3_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
27
|
Cao H, Zhang YQ. Spinal glial activation contributes to pathological pain states. Neurosci Biobehav Rev 2008; 32:972-983. [PMID: 18471878 DOI: 10.1016/j.neubiorev.2008.03.009] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 02/28/2008] [Accepted: 03/13/2008] [Indexed: 12/21/2022]
Abstract
Chronic pain, a pathological state, affects millions of people worldwide. Despite decades of study on the neuronal processing of pain, mechanisms underlying the creation and maintenance of enhanced pain states after injury or inflammation remain far from clear. In the last decade, however, the discovery that glial activation amplifies pain has challenged classic neuronal views of "pain". This review focuses on recent developments in understanding that spinal cord glia are involved in pathological pain. We overview the action of spinal glia (both microglia and astrocytes) in several persistent pain models, and provide new evidence that spinal glia activation contributes to the development and maintenance of arthritic pain facilitation. We also attempt to discuss some critical questions, such as how signals are conveyed from primary afferents to spinal glia following peripheral nerve injury and inflammation. What causes glia to become activated after peripheral/central injury/inflammation? And how the activated glia alter neuronal sensitivity and pain processing? Answers to these questions might open a new approach for treatment of pathological pain.
Collapse
Affiliation(s)
- Hong Cao
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | | |
Collapse
|
28
|
Romero-Sandoval A, Eisenach JC. Clonidine reduces hypersensitivity and alters the balance of pro- and anti-inflammatory leukocytes after local injection at the site of inflammatory neuritis. Brain Behav Immun 2007; 21:569-80. [PMID: 17079113 PMCID: PMC1949871 DOI: 10.1016/j.bbi.2006.09.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 09/01/2006] [Accepted: 09/02/2006] [Indexed: 01/08/2023] Open
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.
Collapse
Affiliation(s)
- Alfonso Romero-Sandoval
- Department of Anesthesiology and Center for the Study of Pharmacologic Plasticity in the Presence of Pain, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1009, USA
| | | |
Collapse
|
29
|
Romero-Sandoval EA, McCall C, Eisenach JC. Alpha2-adrenoceptor stimulation transforms immune responses in neuritis and blocks neuritis-induced pain. J Neurosci 2006; 25:8988-94. [PMID: 16192389 PMCID: PMC6725591 DOI: 10.1523/jneurosci.2995-05.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neuropathic pain may be primarily driven by immune responses in peripheral nerves. Peripherally released catecholamines may exacerbate neuropathic pain and also modulate immune responses in a complex and sometimes opposing manner by actions on multiple adrenoceptor subtypes. We showed previously that injection of the alpha2-adrenoceptor agonist clonidine at the site of peripheral nerve injury reduces pain behavior and local tissue pro-inflammatory cytokine content in rats. The current study used a model of acute inflammatory neuritis to test the efficacy and mechanisms of action of alpha2-adrenoceptor stimulation to reduce pain. Zymosan, injected on the sciatic nerve, caused hypersensitivity to mechanical stimuli ipsilateral to injection and contralaterally, so-called mirror image pain. Ipsilateral hypersensitivity was inhibited dose-dependently by perineural injection of clonidine. Zymosan increased leukocyte number at the site of injection 3 d later as well as their content of interleukin 1alpha (IL-1alpha), IL-1beta, and IL-6. Perineural clonidine prevented both the increase in leukocyte number and cytokine expression induced by zymosan. Additionally, clonidine reduced the capacity of leukocytes to express pro-inflammatory cytokines as assessed by treatment of cells ex vivo with lipopolysaccharide, whereas no repression of IL-10 production occurred. Clonidine reduced the number of macrophages and lymphocytes as well as their expression of tumor necrosis factor alpha. All of the effects of clonidine were prevented by coadministration of an alpha2A-adrenoceptor-preferring antagonist. These results suggest that alpha2-adrenoceptor stimulation transforms cytokine gene expression, especially in macrophages and lymphocytes from a pro- to an anti-inflammatory profile in the setting of neuritis, likely relieving neuritis-induced pain by this mechanism.
Collapse
Affiliation(s)
- E Alfonso Romero-Sandoval
- Department of Anesthesiology, Center for the Study of Pharmacologic Plasticity in the Presence of Pain, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1009, USA
| | | | | |
Collapse
|
30
|
Nee RJ, Butler D. Management of peripheral neuropathic pain: Integrating neurobiology, neurodynamics, and clinical evidence. Phys Ther Sport 2006. [DOI: 10.1016/j.ptsp.2005.10.002] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
31
|
|
32
|
|
33
|
Ledeboer A, Gamanos M, Lai W, Martin D, Maier SF, Watkins LR, Quan N. Involvement of spinal cord nuclear factor κB activation in rat models of proinflammatory cytokine-mediated pain facilitation. Eur J Neurosci 2005; 22:1977-86. [PMID: 16262636 DOI: 10.1111/j.1460-9568.2005.04379.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proinflammatory cytokines, such as interleukin-1beta and tumour necrosis factor-alpha, are released by activated glial cells in the spinal cord and play a major role in pain facilitation. These cytokines exert their actions, at least partially, through the activation of the transcription factor, nuclear factor kappaB (NF-kappaB). In turn, NF-kappaB regulates the transcription of many inflammatory mediators, including cytokines. We have previously shown that intrathecal injection of the human immunodeficiency virus-1 (HIV-1) envelope glycoprotein, gp120, induces mechanical allodynia via the release of proinflammatory cytokines. Here, we investigated whether NF-kappaB is involved in gp120-induced pain behaviour in Sprague-Dawley rats. Intrathecal administration of NF-kappaB inhibitors, pyrrolidinedithiocarbamate (PDTC) and SN50, prior to gp120 partially attenuated gp120-induced allodynia. In addition, PDTC delayed and reversed allodynia in a model of neuropathic pain induced by sciatic nerve inflammation. These observations suggest that intrathecal gp120 may lead to activation of NF-kappaB within the spinal cord. To reveal NF-kappaB activation, we assessed inhibitory factor kappaBalpha (IkappaBalpha) mRNA expression by in situ hybridization, as NF-kappaB activation up-regulates IkappaBalpha gene expression as part of an autoregulatory feedback loop. No or low levels of IkappaBalpha mRNA were detected in the lumbar spinal cord of vehicle-injected rats, whereas IkappaBalpha mRNA expression was markedly induced in the spinal cord following intrathecal gp120 in predominantly astrocytes and endothelial cells. Moreover, IkappaBalpha mRNA expression positively correlated with proinflammatory cytokine protein levels in lumbosacral cerebrospinal fluid. Together, these results demonstrate that spinal cord NF-kappaB activation is involved, at least in part, in exaggerated pain states.
Collapse
Affiliation(s)
- Annemarie Ledeboer
- Department of Psychology & Center for Neuroscience, University of Colorado at Boulder, Boulder, CO 80309-0345, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Zelenka M, Schäfers M, Sommer C. Intraneural injection of interleukin-1beta and tumor necrosis factor-alpha into rat sciatic nerve at physiological doses induces signs of neuropathic pain. Pain 2005; 116:257-263. [PMID: 15964142 DOI: 10.1016/j.pain.2005.04.018] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2004] [Revised: 03/05/2005] [Accepted: 04/18/2005] [Indexed: 10/25/2022]
Abstract
Proinflammatory cytokines are mediators of inflammatory and neuropathic pain. Here, we investigated pain-related behavior in rats after intraneural injection of different doses of rat recombinant interleukin-1beta (rrIL-1beta) and tumor necrosis factor-alpha (rrTNF) into the sciatic nerve. Doses ranged between 0.25 and 2500pg/ml for rrIL-1beta and 0.25-250pg/ml for rrTNF. Thermal hyperalgesia as measured according to the Hargreaves method was most prominent with 2.5pg/ml of rrIL-1beta or rrTNF. Mechanical allodynia as assessed using von Frey hairs was seen consistently with 2.5pg/ml of rrIL-1beta and 0.25-2.5pg/ml of rrTNF. Higher and lower doses had no significant effect on pain-related behavior. Morphometric analysis of semithin sections of the sciatic nerve 10 days after the injections revealed no significant fiber loss. The fiber size distribution was not significantly altered by any of the treatments. Particularly with injections of rrIL-1beta, an increase of epineurial macrophages was observed at all doses. The immunohistochemical expression of cellular markers of neuronal damage (activating transcription factor 3) or activation (phosphorylated p38 mitogen-activated kinase, NF-kappa B p65) in dorsal root ganglia (DRG) tended to increase with both cytokine injections. However, this did not reflect the extent of behavioral changes. In summary, we found a bell-shaped dose-response curve for the algesic effects of rrIL-1beta and rrTNF, peaking at doses equivalent to those of endogenous cytokines released locally after nerve injury. The absence of corresponding morphological changes in nerves supports the concept of a functional effect of the cytokines at these doses.
Collapse
Affiliation(s)
- Marek Zelenka
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, D-97080 Würzburg, Germany
| | | | | |
Collapse
|
35
|
Milligan ED, Langer SJ, Sloane EM, He L, Wieseler-Frank J, O'Connor K, Martin D, Forsayeth JR, Maier SF, Johnson K, Chavez RA, Leinwand LA, Watkins LR. Controlling pathological pain by adenovirally driven spinal production of the anti-inflammatory cytokine, interleukin-10. Eur J Neurosci 2005; 21:2136-48. [PMID: 15869510 DOI: 10.1111/j.1460-9568.2005.04057.x] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gene therapy for the control of pain has, to date, targeted neurons. However, recent evidence supports that spinal cord glia are critical to the creation and maintenance of pain facilitation through the release of proinflammatory cytokines. Because of the ability of interleukin-10 (IL-10) to suppress proinflammatory cytokines, we tested whether an adenoviral vector encoding human IL-10 (AD-h-IL10) would block and reverse pain facilitation. Three pain models were examined, all of which are mediated by spinal pro-inflammatory cytokines. Acute intrathecal administration of rat IL-10 protein itself briefly reversed chronic constriction injury-induced mechanical allodynia and thermal hyperalgesia. The transient reversal caused by IL-10 protein paralleled the half-life of human IL-10 protein in the intrathecal space (t(1/2) approximately 2 h). IL-10 gene therapy both prevented and reversed thermal hyperalgesia and mechanical allodynia, without affecting basal responses to thermal or mechanical stimuli. Extra-territorial, as well as territorial, pain changes were reversed by this treatment. Intrathecal AD-h-IL10 injected over lumbosacral spinal cord led to elevated lumbosacral cerebrospinal fluid (CSF) levels of human IL-10, with far less human IL-10 observed in cervical CSF. In keeping with IL-10's known anti-inflammatory actions, AD-h-IL10 lowered CSF levels of IL-1, relative to control AD. These studies support that this gene therapy approach provides an alternative to neuronally focused drug and gene therapies for clinical pain control.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ledeboer A, Sloane EM, Milligan ED, Frank MG, Mahony JH, Maier SF, Watkins LR. Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain 2005; 115:71-83. [PMID: 15836971 DOI: 10.1016/j.pain.2005.02.009] [Citation(s) in RCA: 543] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Revised: 12/06/2004] [Accepted: 02/07/2005] [Indexed: 01/23/2023]
Abstract
Activated glial cells (microglia and astroglia) in the spinal cord play a major role in mediating enhanced pain states by releasing proinflammatory cytokines and other substances thought to facilitate pain transmission. In the present study, we report that intrathecal administration of minocycline, a selective inhibitor of microglial cell activation, inhibits low threshold mechanical allodynia, as measured by the von Frey test, in two models of pain facilitation. In a rat model of neuropathic pain induced by sciatic nerve inflammation (sciatic inflammatory neuropathy, SIN), minocycline delayed the induction of allodynia in both acute and persistent paradigms. Moreover, minocycline was able to attenuate established SIN-induced allodynia 1 day, but not 1 week later, suggesting a limited role of microglial activation in more perseverative pain states. Our data are consistent with a crucial role for microglial cells in initiating, rather than maintaining, enhanced pain responses. In a model of spinal immune activation by intrathecal HIV-1 gp120, we show that the anti-allodynic effects of minocycline are associated with decreased microglial activation, attenuated mRNA expression of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), IL-1beta-converting enzyme, TNF-alpha-converting enzyme, IL-1 receptor antagonist and IL-10 in lumbar dorsal spinal cord, and reduced IL-1beta and TNF-alpha levels in the CSF. In contrast, no significant effects of minocycline were observed on gp120-induced IL-6 and cyclooxygenase-2 expression in spinal cord or CSF IL-6 levels. Taken together these data highlight the importance of microglial activation in the development of exaggerated pain states.
Collapse
Affiliation(s)
- Annemarie Ledeboer
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Campus Box 345, Boulder, CO 80309-0345, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Twining CM, Sloane EM, Schoeniger DK, Milligan ED, Martin D, Marsh H, Maier SF, Watkins LR. Activation of the spinal cord complement cascade might contribute to mechanical allodynia induced by three animal models of spinal sensitization. THE JOURNAL OF PAIN 2005; 6:174-83. [PMID: 15772911 DOI: 10.1016/j.jpain.2004.11.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The present series of experiments examined whether the complement cascade might play a key role in the expression of mechanical allodynia. Soluble complement receptor 1 (sCR1) was used to block the activation of the membrane attack pathway of the complement cascade. In doing so, sCR1 prevents the formation of the biologically active end products C3a, C5a, and membrane attack complexes (MACs). Intrathecal sCR1 had no effect on the behavioral responses of control groups. In contrast, blockade of this pathway abolished the expression of mechanical allodynia induced by peripheral nerve inflammation (sciatic inflammatory neuropathy model), partial sciatic nerve injury (chronic constriction injury model), and intrathecal injection of human immunodeficiency virus type 1 gp120, a viral envelope protein that activates glia. The fact that enhanced nociception was prevented or reversed in all 3 paradigms suggests that complement might be broadly involved in spinally mediated pain enhancement. The mechanisms whereby complement activation might potentially affect the functioning of microglia, astrocytes, and neurons are discussed. The complement cascade has not been previously implicated in spinal sensitization. These data suggest that complement activation within the spinal cord might contribute to enhanced pain states and provide additional evidence for immune regulation of pain transmission.
Collapse
Affiliation(s)
- Carin M Twining
- Department of Psychology and the Center of Neuroscience, University of Colorado at Boulder, 80309, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Milligan ED, Sloane EM, Langer SJ, Cruz PE, Chacur M, Spataro L, Wieseler-Frank J, Hammack SE, Maier SF, Flotte TR, Forsayeth JR, Leinwand LA, Chavez R, Watkins LR. Controlling neuropathic pain by adeno-associated virus driven production of the anti-inflammatory cytokine, interleukin-10. Mol Pain 2005; 1:9. [PMID: 15813997 PMCID: PMC1079940 DOI: 10.1186/1744-8069-1-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2005] [Accepted: 02/25/2005] [Indexed: 01/06/2023] Open
Abstract
Despite many decades of drug development, effective therapies for neuropathic pain remain elusive. The recent recognition of spinal cord glia and glial pro-inflammatory cytokines as important contributors to neuropathic pain suggests an alternative therapeutic strategy; that is, targeting glial activation or its downstream consequences. While several glial-selective drugs have been successful in controlling neuropathic pain in animal models, none are optimal for human use. Thus the aim of the present studies was to explore a novel approach for controlling neuropathic pain. Here, an adeno-associated viral (serotype II; AAV2) vector was created that encodes the anti-inflammatory cytokine, interleukin-10 (IL-10). This anti-inflammatory cytokine is known to suppress the production of pro-inflammatory cytokines. Upon intrathecal administration, this novel AAV2-IL-10 vector was successful in transiently preventing and reversing neuropathic pain. Intrathecal administration of an AAV2 vector encoding beta-galactosidase revealed that AAV2 preferentially infects meningeal cells surrounding the CSF space. Taken together, these data provide initial support that intrathecal gene therapy to drive the production of IL-10 may prove to be an efficacious treatment for neuropathic pain.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Evan M Sloane
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Stephen J Langer
- Department of Molecular, Cellular & Developmental Biology, University of CO at Boulder, Boulder, CO 80309 USA
| | - Pedro E Cruz
- Genetics Institute, the Powell Gene Therapy Center & Department of Pediatrics, University of FL at Gainesville, Gainesville, FL 32610 USA
| | - Marucia Chacur
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Leah Spataro
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Julie Wieseler-Frank
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Sayamwong E Hammack
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Steven F Maier
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Terence R Flotte
- Genetics Institute, the Powell Gene Therapy Center & Department of Pediatrics, University of FL at Gainesville, Gainesville, FL 32610 USA
| | | | - Leslie A Leinwand
- Department of Molecular, Cellular & Developmental Biology, University of CO at Boulder, Boulder, CO 80309 USA
| | | | - Linda R Watkins
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| |
Collapse
|
39
|
Abstract
Whereas small-fibre sensory neuropathies might ultimately lead to cell death and loss of sensation, they first progress through a phase, which might last for years, characterized by the presence of analgesia-resistant neuropathic dysesthesias and pain. Much previous research has addressed these two phases as separate phenomena mediated by presumably discrete biochemical mechanisms. We hypothesized that activity in signalling pathways that ultimately lead to apoptosis plays a critical role in the generation of neuropathic pain, before death of sensory neurons becomes apparent. We have tested the hypothesis that activator and effector caspases, defining components of programmed cell death (apoptosis) signalling pathways, also contribute to pain-related behaviour in animals with small-fibre peripheral neuropathies and that the death receptor ligand, tumour necrosis factor-alpha, and its downstream second messenger, ceramide, also produce pain-related behaviour via this mechanism. In two models of painful peripheral neuropathy, HIV/AIDS therapy (induced by the nucleoside reverse transcriptase inhibitor, dideoxycytidine), and cancer chemotherapy (induced by vincristine) peripheral neuropathy, and for pain-related behaviour induced by tumour necrosis factor-alpha and its second messenger, ceramide, inhibition of both activator (1, 2, 8 and 9) and effector (3) caspases attenuates neuropathic pain-related behaviour, although has no effect in streptozotocin-diabetic neuropathy and control rats. We conclude that during a latent phase, before apoptotic cell death is manifest, the caspase signalling pathway can contribute to pain in small-fibre peripheral neuropathies, and that inflammatory/immune mediators also activate these pathways. This suggests that these pathways are potential targets for novel pharmacological agents for the treatment of inflammatory as well as neuropathic pain.
Collapse
Affiliation(s)
- Elizabeth K Joseph
- Department of Medicine, Division of Neuroscience and Biomedical Sciences Programme, NIH Pain Centre, Box # 0440/C522, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | |
Collapse
|
40
|
|
41
|
Verge GM, Milligan ED, Maier SF, Watkins LR, Naeve GS, Foster AC. Fractalkine (CX3CL1) and fractalkine receptor (CX3CR1) distribution in spinal cord and dorsal root ganglia under basal and neuropathic pain conditions. Eur J Neurosci 2004; 20:1150-60. [PMID: 15341587 DOI: 10.1111/j.1460-9568.2004.03593.x] [Citation(s) in RCA: 301] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fractalkine is a unique chemokine reported to be constitutively expressed by neurons. Its only receptor, CX3CR1, is expressed by microglia. Little is known about the expression of fractalkine and CX3CR1 in spinal cord. Given that peripheral nerve inflammation and/or injury gives rise to neuropathic pain, and neuropathic pain may be partially mediated by spinal cord glial activation and consequent glial proinflammatory cytokine release, there must be a signal released by affected neurons that triggers the activation of glia. We sought to determine whether there is anatomical evidence implicating spinal fractalkine as such a neuron-to-glia signal. We mapped the regional and cellular localization of fractalkine and CX3CR1 in the rat spinal cord and dorsal root ganglion, under basal conditions and following induction of neuropathic pain, employing both an inflammatory (sciatic inflammatory neuropathy; SIN) as well as a traumatic (chronic constriction injury; CCI) model. Fractalkine immunoreactivity and mRNA were observed in neurons, but not glia, in the rat spinal cord and dorsal root ganglia, and levels did not change following either CCI or SIN. By contrast, CX3CR1 was expressed by microglia in the basal state, and the microglial cellular concentration was up-regulated in a regionally specific manner in response to neuropathy. CX3CR1-expressing cells were identified as microglia by their cellular morphology and positive OX-42 and CD4 immunostaining. The cellular distribution of fractalkine and CX3CR1 in the spinal circuit associated with nociceptive transmission supports a potential role in the mechanisms that contribute to the exaggerated pain state in these models of neuropathy.
Collapse
Affiliation(s)
- Gail M Verge
- Neurocrine Biosciences, 12790 El Camino Real, San Diego, CA 92130, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Twining CM, Sloane EM, Milligan ED, Chacur M, Martin D, Poole S, Marsh H, Maier SF, Watkins LR. Peri-sciatic proinflammatory cytokines, reactive oxygen species, and complement induce mirror-image neuropathic pain in rats. Pain 2004; 110:299-309. [PMID: 15275780 DOI: 10.1016/j.pain.2004.04.008] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2003] [Revised: 03/25/2004] [Accepted: 04/05/2004] [Indexed: 11/29/2022]
Abstract
In inflammatory neuropathy, immune activation near intact peripheral nerves induces mechanical allodynia. The identity of the peripheral immune product(s) that lead to these changes in pain behavior is unknown. The present series of studies utilized the sciatic inflammatory neuropathy (SIN) model to examine this question. Here, inflammatory neuropathy is created by injecting an immune activator (zymosan) around one sciatic nerve via an indwelling catheter. Our prior studies demonstrated that peri-sciatic zymosan activated macrophages and neutrophils to release proinflammatory cytokines and reactive oxygen species (ROS). In addition, zymosan is a classical activator of the complement cascade. Thus the present series of experiments examined whether any of these inflammatory mediators are involved in the initial induction of SIN-induced ipsilateral or bilateral allodynias. Peri-sciatic injection of selective inhibitors/antagonists revealed that a number of immune products are early mediators of the resultant allodynias, including proinflammatory cytokines (tumor necrosis factor, interleukin-1, and interleukin-6), ROS, and complement. Thus these immune-derived substances can markedly alter sensory nerve function at mid-axon.
Collapse
Affiliation(s)
- Carin M Twining
- Department of Psychology and the Center for Neuroscience, University of Colorado at Boulder, Campus Box 345. Boulder, CO 80309-0345, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Milligan ED, Zapata V, Chacur M, Schoeniger D, Biedenkapp J, O'Connor KA, Verge GM, Chapman G, Green P, Foster AC, Naeve GS, Maier SF, Watkins LR. Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats. Eur J Neurosci 2004; 20:2294-302. [PMID: 15525271 DOI: 10.1111/j.1460-9568.2004.03709.x] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent evidence suggests that spinal cord glia can contribute to enhanced nociceptive responses. However, the signals that cause glial activation are unknown. Fractalkine (CX3C ligand-1; CX3CL1) is a unique chemokine expressed on the extracellular surface of spinal neurons and spinal sensory afferents. In the dorsal spinal cord, fractalkine receptors are primarily expressed by microglia. As fractalkine can be released from neurons upon strong activation, it has previously been suggested to be a neuron-to-glial signal that induces glial activation. The present series of experiments provide an initial investigation of the spinal pain modulatory effects of fractalkine. Intrathecal fractalkine produced dose-dependent mechanical allodynia and thermal hyperalgesia. In addition, a single injection of fractalkine receptor antagonist (neutralizing antibody against rat CX3C receptor-1; CX3CR1) delayed the development of mechanical allodynia and/or thermal hyperalgesia in two neuropathic pain models: chronic constriction injury (CCI) and sciatic inflammatory neuropathy. Intriguingly, anti-CX3CR1 reduced nociceptive responses when administered 5-7 days after CCI, suggesting that prolonged release of fractalkine may contribute to the maintenance of neuropathic pain. Taken together, these initial investigations of spinal fractalkine effects suggest that exogenous and endogenous fractalkine are involved in spinal sensitization, including that induced by peripheral neuropathy.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- CX3C Chemokine Receptor 1
- Cell Communication/drug effects
- Cell Communication/physiology
- Chemokine CX3CL1
- Chemokines, CX3C/administration & dosage
- Chemokines, CX3C/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Hyperalgesia/chemically induced
- Hyperalgesia/metabolism
- Hyperalgesia/physiopathology
- Injections, Spinal
- Ligation
- Male
- Membrane Proteins/administration & dosage
- Membrane Proteins/drug effects
- Membrane Proteins/metabolism
- Microglia/drug effects
- Microglia/metabolism
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neurons/metabolism
- Nociceptors/drug effects
- Nociceptors/physiology
- Pain/chemically induced
- Pain/metabolism
- Pain/physiopathology
- Peripheral Nervous System Diseases/chemically induced
- Peripheral Nervous System Diseases/metabolism
- Peripheral Nervous System Diseases/physiopathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytokine/antagonists & inhibitors
- Receptors, Cytokine/metabolism
- Receptors, HIV/antagonists & inhibitors
- Receptors, HIV/metabolism
- Sciatic Neuropathy/chemically induced
- Sciatic Neuropathy/metabolism
- Sciatic Neuropathy/physiopathology
- Spinal Cord/drug effects
- Spinal Cord/metabolism
- Spinal Cord/physiopathology
Collapse
Affiliation(s)
- E D Milligan
- Department of Psychology & the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO 80309-0345, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Eliav E, Tal M, Benoliel R. Experimental malignancy in the rat induces early hypersensitivity indicative of neuritis. Pain 2004; 110:727-737. [PMID: 15288414 DOI: 10.1016/j.pain.2004.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Revised: 04/16/2004] [Accepted: 05/12/2004] [Indexed: 11/26/2022]
Abstract
Cancer pain mechanisms involve multiple factors including the accompanying inflammatory process neural effects. Inflammation along a nerve trunk (neuritis) has been shown to induce hypersensitivity at the innervated target organ. In this experiment the neural effects of MAT B mammary adenocarcinoma cells implanted adjacent to sciatic nerve (MAT group) were compared to those induced by thymus cell extract (THY), saline (SAL) and the potent proinflammatory agent Complete Freund's adjuvant (CFA). Significant pain behavior was detected only in the ipsilateral hindpaw of MAT and CFA rats lasting seven days post-operatively (dpo). On the ninth dpo MAT rats developed significant hyposensitivity to mechanical and electrical stimuli. Interleukin (IL)-6 levels (ELISA) from MAT and CFA exposed nerves were significantly elevated at dpo 2 and remained so in MAT at dpo 8. Indomethacin (1 mg/kg i.p.) abolished the observed pain responses in MAT and CFA rats exposed nerves. Light microscopy of the MAT nerves at the second dpo revealed neural infiltration of immune and malignant cells with mild edema. By the seventh dpo there was nerve damage and at dpo 14 nerve tissue was largely replaced by malignant and immune cells. Electrophysiology of saphenous nerves exposed to MAT, CFA or SAL revealed significantly increased spontaneous activity in MAT and CFA groups. Spike response to hindpaw mechanical stimulation was significantly reduced only in the MAT group (dpo 6-9) suggestive of nerve damage. Inflammatory neuritis is an early expression of malignancy and may play a role in chronic cancer-related pain initiation and additionally may offer diagnostic opportunities.
Collapse
Affiliation(s)
- Eli Eliav
- Robert & Susan Carmel Endowed Chair in Algesiology UMDNJ-New Jersey Dental School, 110 Bergen Street, Newark, NJ 07103, USA Department of Anatomy and Cell Biology, Faculty of Dental Medicine, The Hebrew University-Hadassah, P.O. Box 12272, Jerusalem 91120, Israel Department of Oral Medicine, Faculty of Dental Medicine, The Hebrew University-Hadassah, P.O. Box 12272, Jerusalem 91120, Israel
| | | | | |
Collapse
|
45
|
Walker SJ, Worst TJ, Freeman WM, Vrana KE. Functional genomic analysis in pain research using hybridization arrays. METHODS IN MOLECULAR MEDICINE 2004; 99:239-53. [PMID: 15131342 DOI: 10.1385/1-59259-770-x:239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Hybridization array technology makes it possible to compare global gene-expression patterns in any experimental context for which good-quality RNA can be generated. To date, DNA arrays have been used as a tool to compare functional genomic changes (differences in wholesale gene expression) in studies that cover an impressive variety of research disciplines including cancer, yeast genomics, and, more recently, neuroscience and behavior. The basic premise of the array experiment is that one interrogates a panel of probes (gene-specific cDNA fragments or gene-specific oligonucleotides that have been immobilized on a solid support) with RNAs (targets) from control and treated experimental samples that have been either radioactively or fluorescently labeled. Signal derived from either competitive (both samples on a single chip) or differential (one sample/one chip) hybridization is used to calculate relative gene expression. There are three widely used platforms available to perform array experiments (Affymetrix GeneChips, oligonucleotide arrays, and membrane-based cDNA arrays) and each platform offers advantages and limitations. The experimental description in this chapter explains, in detail, how to perform a hybridization array using the macroarray platform.
Collapse
Affiliation(s)
- Stephen J Walker
- Center for the Neurobiological Investigation of Drug Abuse and Neurobehavioral Study of Alcohol, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | | |
Collapse
|
46
|
Chacur M, Milligan ED, Sloan EM, Wieseler-Frank J, Barrientos RM, Martin D, Poole S, Lomonte B, Gutiérrez JM, Maier SF, Cury Y, Watkins LR. Snake venom phospholipase A2s (Asp49 and Lys49) induce mechanical allodynia upon peri-sciatic administration: involvement of spinal cord glia, proinflammatory cytokines and nitric oxide. Pain 2004; 108:180-91. [PMID: 15109522 DOI: 10.1016/j.pain.2003.12.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Revised: 12/10/2003] [Accepted: 12/18/2003] [Indexed: 10/26/2022]
Abstract
Snakebites constitute a serious public health problem in Central and South America, where species of the lancehead pit vipers (genus Bothrops) cause the majority of accidents. Bothrops envenomations are very painful, and this effect is not neutralized by antivenom treatment. Two variants of secretory phospholipases A2 (sPLA2), corresponding to Asp49 and Lys49 PLA2s, have been isolated from Bothrops asper venom. These sPLA2s induce hyperalgesia in rats following subcutaneous injection. However, venom in natural Bothrops bites is frequently delivered intramuscularly, thereby potentially reaching peripheral nerve bundles. Thus, the present series of experiments tested whether these sPLA2s could exert pain-enhancing effects following administration around healthy sciatic nerve. Both were found to produce mechanical allodynia ipsilateral to the injection site; no thermal hyperalgesia was observed. As no prior study has examined potential spinal mechanisms underlying sPLA2 actions, a series of anatomical and pharmacological studies were performed. These demonstrated that both sPLA2s produce activation of dorsal horn astrocytes and microglia that is more prominent ipsilateral to the site of injection. As proinflammatory cytokines and nitric oxide have each been previously implicated in spinally mediated pain facilitation, the effect of pharmacological blockade of these substances was tested. The results demonstrate that mechanical allodynia induced by both sPLA2s is blocked by interleukin-1 receptor antagonist, anti-rat interleukin-6 neutralizing antibody, the anti-inflammatory cytokine interleukin-10, and a nitric oxide synthesis inhibitor (L-NAME). As a variety of immune cells also produce and release sPLA2s during inflammatory states, the data may have general implications for the understanding of inflammatory pain.
Collapse
|
47
|
Abstract
There are no known anatomical connections between neurons that innervate homologous right and left body parts. Nevertheless, some patients develop bilateral abnormalities after unilateral injury, a phenomenon often unrecognized and not yet characterized. Therefore, we examined in rats the effects of ligating and cutting one tibial nerve on sensory function and on density of innervation in hind paws contralaterally as well as ipsilaterally to the injury, at times between 1 day and 5 months after surgery. Punches removed from tibial- or sural-innervated planter paw skin were immunolabeled to quantitate epidermal nerve endings. Naive and sham-operated rats provided controls. Axotomized rats had near-total loss of PGP9.5(+) innervation within ipsilateral tibial-innervated skin at all time-points. Adjacent ipsilateral sural-innervated skin had persistent hyperalgesia without denervation, and robust axonal sprouting at 5 months after surgery. Contralesional hind paws lost 54% of innervation in tibial-innervated epidermis starting 1 week after surgery and persisting throughout. Contralesional sural-innervated skin had neither neurite loss nor sprouting. These results imply that unilateral nerve injury can cause profound, long lasting, nerve-branch-specific loss of distal innervation contralaterally as well as ipsilaterally. They discredit the practice of using tissues contralateral to an injury to provide normative controls and suggest the possibility of rapid, transmedian postinjury signals between homologous mirror-image neurons.
Collapse
Affiliation(s)
- Anne Louise Oaklander
- Nerve Injury Unit, Department of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | | |
Collapse
|
48
|
McCluskey LP. Up-regulation of activated macrophages in response to degeneration in the taste system: Effects of dietary sodium restriction. J Comp Neurol 2004; 479:43-55. [PMID: 15389612 DOI: 10.1002/cne.20307] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dietary sodium restriction combined with unilateral chorda tympani nerve section leads to a rapid and specific decrease in neurophysiological taste responses to sodium in the contralateral, intact chorda tympani (Hill and Phillips [1994] J. Neurosci. 14:2904-2910). Previous work demonstrated that dietary sodium restriction may induce these early functional deficits by inhibiting immune activity after denervation (Phillips and Hill [1996] Am. J. Physiol. 271:R857-R862). However, little is known about the leukocyte response to denervation of taste buds in fungiform papillae. In the current study, it was hypothesized that T cells and macrophages are increased in the tongue after unilateral denervation in control-fed but not sodium-restricted animals. Adult, specified pathogen-free rats received unilateral chorda tympani nerve section or sham section followed by dietary sodium restriction or maintenance on control diet. At day 1, 2, 5, 7, or 50 postsectioning, immunostaining was used to detect the percentage of staining for activated macrophages, the number of alpha beta T cells, and the number of delta gamma epithelial T cells in the tongue. The number of lingual T cells did not significantly differ between treatment groups following denervation. However, there was a dramatic bilateral increase in ED1(+) staining for activated macrophages in control-fed rats that peaked at day 2 postsectioning. In contrast, sodium-restricted rats did not show an increase in activated macrophages above baseline at any time postsectioning. Further analysis of extralingual macrophages indicated that the deficit in immune activity in sodium-restricted rats is localized to the tongue and is not widespread. A model for immune modulation of taste receptor cell function is proposed based on these novel findings.
Collapse
|
49
|
Lee DH, Iyengar S, Lodge D. The role of uninjured nerve in spinal nerve ligated rats points to an improved animal model of neuropathic pain. Eur J Pain 2003; 7:473-9. [PMID: 12935800 DOI: 10.1016/s1090-3801(03)00019-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
L5 and L6 spinal nerve ligation (SNL) in rats leads to behavioral signs of neuropathic pain including mechanical allodynia. The purposes of this study were to investigate the role of the intact L4 spinal nerve in the development of mechanical allodynia following L5 and L6 SNL and, as a result, to develop a modified model of neuropathic pain. As a first set of experiments, in addition to tight ligation of the left L5 and L6 spinal nerves, the intact L4 spinal nerve was manipulated either (1) by gentle repeated stretching of the L4 spinal nerve immediately after L5 and L6 SNL or (2) by intermittent mechanical stimulation to the ipsilateral paw during the first week after SNL. Tactile sensitivity was measured by determining the foot withdrawal threshold before and after SNL. Mild irritation of L4 spinal nerve and application of mechanical stimuli to the ipsilateral paw significantly increased the development of mechanical allodynia after SNL. In a second set of experiments, SNL was produced by tightly ligating only the left L5 spinal nerve with or without a loop of 5-0 chromic gut placed loosely around the L4 spinal nerve. This additional L4 loop significantly increased long-lasting tactile sensitivity compared to L5 SNL alone. These results suggest that afferent activity of the intact L4 spinal nerve aids in the development of mechanical allodynia in the SNL model of neuropathic pain. The addition of a chromic gut loop around the intact L4 spinal nerve can augment the development of mechanical allodynia following SNL of L5. We propose this latter as a useful and practical animal model of neuropathic pain.
Collapse
Affiliation(s)
- Doo H Lee
- Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | |
Collapse
|
50
|
Abstract
Chronic pain, especially neuropathic pain and cancer pain, is often not adequately treated by currently available analgesics. Animal models provide pivotal systems for preclinical study of pain. This article reviews some of the most widely used or promising new models for chronic pain. Partial spinal ligation, chronic constriction injury, and L5/L6 spinal nerve ligation represent three of the best characterized rodent models of peripheral neuropathy. Recently, several mouse and rat bone cancer pain models have been reported. Primary or permanent cultures of sensory neurons have been established to study the molecular mechanism of pain, especially for neurotransmitter release and signal transduction. The emerging gene microarray, genomics and proteomics methods may be applied to throughly characterize these cells. Each model is uniquely created with distinct mechanisms, it is therefore essential to report and interpret results in the context of a specific model.
Collapse
Affiliation(s)
- Lili X Wang
- Department of Biopharmaceutical Sciences, University of Illinois, 833 South Woods Street, Chicago, IL 60612, USA
| | | |
Collapse
|