1
|
Motzkin JC, Basbaum AI, Crowther AJ. Neuroanatomy of the nociceptive system: From nociceptors to brain networks. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:1-39. [PMID: 39580210 DOI: 10.1016/bs.irn.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter reviews the neuroanatomy of the nociceptive system and its functional organization. We describe three main compartments of the nervous system that underlie normal nociception and the resulting pain percept: Peripheral, Spinal Cord, and Brain. We focus on how ascending nociceptive processing streams traverse these anatomical compartments, culminating in the multidimensional experience of pain. We also describe neuropathic pain conditions, in which nociceptive processing is abnormal, not only because of the primary effects of a lesion or disease affecting peripheral nerves or the central nervous system (CNS), but also due to secondary effects on ascending pathways and brain networks. We discuss how the anatomical components (circuits/networks) reorganize under various etiologies of neuropathic pain and how these changes can give rise to pathological pain states.
Collapse
Affiliation(s)
- Julian C Motzkin
- Department of Neurology and Department Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States.
| | - Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Andrew J Crowther
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
2
|
Abboud C, Duveau A, Bouali-Benazzouz R, Massé K, Mattar J, Brochoire L, Fossat P, Boué-Grabot E, Hleihel W, Landry M. Animal models of pain: Diversity and benefits. J Neurosci Methods 2020; 348:108997. [PMID: 33188801 DOI: 10.1016/j.jneumeth.2020.108997] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions. In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.
Collapse
Affiliation(s)
- Cynthia Abboud
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Alexia Duveau
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Rabia Bouali-Benazzouz
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Karine Massé
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Joseph Mattar
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Louison Brochoire
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Pascal Fossat
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Eric Boué-Grabot
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Walid Hleihel
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Lebanon; Faculty of Arts and Sciences, Holy Spirit University of Kaslik (USEK), Lebanon
| | - Marc Landry
- Univ. Bordeaux, CNRS, Institute for Neurodegenerative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France.
| |
Collapse
|
3
|
Onishi T, Watanabe T, Sasaki M, Kamiya Y, Horie M, Tsukano H, Hishida R, Kohno T, Takebayashi H, Baba H, Shibuki K. Acute spatial spread of NO-mediated potentiation during hindpaw ischaemia in mice. J Physiol 2019; 597:3441-3455. [PMID: 31087329 PMCID: PMC6851834 DOI: 10.1113/jp277615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/29/2019] [Indexed: 12/31/2022] Open
Abstract
Key points Neuropathic pain spreads spatially beyond the injured sites, and the mechanism underlying the spread has been attributed to inflammation occurring in the spinal cord. However, the spatial spread of spinal/cortical potentiation induced by conduction block of the peripheral nerves can be observed prior to inflammation. In the present study, we found that spreading potentiation and hypersensitivity acutely induced by unilateral hindpaw ischaemia are nitric oxide (NO)‐dependent and that NO is produced by ischaemia and quickly diffuses within the spinal cord. We also found that NO production induced by ischaemia is not observed in the presence of an antagonist for group II metabotropic glutamate receptors (mGluRs) and that neuronal NO synthase‐positive dorsal horn neurons express group II mGluRs. These results suggest strongly that NO‐mediated spreading potentiation in the spinal cord is one of the trigger mechanisms for neuropathic pain.
Abstract Cortical/spinal responses to hindpaw stimulation are bilaterally potentiated by unilateral hindpaw ischaemia in mice. We tested the hypothesis that hindpaw ischaemia produces nitric oxide (NO), which diffuses in the spinal cord to induce spatially spreading potentiation. Using flavoprotein fluorescence imaging, we confirmed that the spreading potentiation in hindpaw responses was induced during ischaemia in the non‐stimulated hindpaw. This spreading potentiation was blocked by spinal application of l‐NAME, an inhibitor of NO synthase (NOS). Furthermore, no spreading potentiation was observed in neural NOS (nNOS) knockout mice. Spinal application of an NO donor was enough to induce cortical potentiation and mechanical hypersensitivity. The spatial distribution of NO during unilateral hindpaw ischaemia was visualized using 4‐amino‐5‐methylamino‐2′,7′‐difluorofluorescein (DAF‐FM). An increase in fluorescence derived from the complex of DAF‐FM with NO was observed on the ischaemic side of the spinal cord. A similar but smaller increase was also observed on the contralateral side. Somatosensory potentiation after hindpaw ischaemia is known to be inhibited by spinal application of LY354740, an agonist of group II metabotropic glutamate receptors (mGluRs). We confirmed that the spinal DAF‐FM fluorescence increases during hindpaw ischaemia were not observed in the presence of LY354740. We also confirmed that approximately half of the nNOS‐positive neurons in the superficial laminae of the dorsal horn expressed mGluR2 mRNA. These results suggest that disinhibition of mGluR2 produces NO which in turn induces a spreading potentiation in a wide area of the spinal cord. Such spreading, along with the consequent non‐specific potentiation in the spinal cord, may trigger neuropathic pain. Neuropathic pain spreads spatially beyond the injured sites, and the mechanism underlying the spread has been attributed to inflammation occurring in the spinal cord. However, the spatial spread of spinal/cortical potentiation induced by conduction block of the peripheral nerves can be observed prior to inflammation. In the present study, we found that spreading potentiation and hypersensitivity acutely induced by unilateral hindpaw ischaemia are nitric oxide (NO)‐dependent and that NO is produced by ischaemia and quickly diffuses within the spinal cord. We also found that NO production induced by ischaemia is not observed in the presence of an antagonist for group II metabotropic glutamate receptors (mGluRs) and that neuronal NO synthase‐positive dorsal horn neurons express group II mGluRs. These results suggest strongly that NO‐mediated spreading potentiation in the spinal cord is one of the trigger mechanisms for neuropathic pain.
Collapse
Affiliation(s)
- Takeshi Onishi
- Department of Neurophysiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan.,Department of Anesthesiology, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Tatsunori Watanabe
- Department of Anesthesiology, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Mika Sasaki
- Department of Anesthesiology, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Yoshinori Kamiya
- Department of Anesthesiology, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Masao Horie
- Department of Morphological Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, 890-8544, Japan
| | - Hiroaki Tsukano
- Department of Neurophysiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Ryuichi Hishida
- Department of Neurophysiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Tatsuro Kohno
- Department of Anesthesiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, 983-8536, Japan
| | - Hirohide Takebayashi
- Department of Neurobiology and Anatomy, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Hiroshi Baba
- Department of Anesthesiology, Faculty of Medicine, Niigata University, Niigata, 951-8510, Japan
| | - Katsuei Shibuki
- Department of Neurophysiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| |
Collapse
|
4
|
Ghanavatian S, Wie CS, Low RS, Butterfield RJ, Zhang N, Dhaliwal GS, Montoya JM, Swanson DL. Parameters associated with efficacy of epidural steroid injections in the management of postherpetic neuralgia: the Mayo Clinic experience. J Pain Res 2019; 12:1279-1286. [PMID: 31118750 PMCID: PMC6503501 DOI: 10.2147/jpr.s190646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/18/2019] [Indexed: 12/04/2022] Open
Abstract
Purpose: Thirty percent of patients with postherpetic neuralgia (PHN) receiving conservative treatment report unsatisfactory pain relief. Epidural steroid injections (ESIs) are commonly used as a therapeutic intervention in these patients. In this study, we aimed to determine if there are variables that predict the efficacy of ESI in patients with PHN. Patients and methods: We retrospectively identified patients seen at Mayo Clinic who had PHN and received ESI. From their medical records, we abstracted the demographic variables, concurrent medication use, anatomic approach and medication for ESI, and degree of pain relief at 2 and 12 weeks' postintervention. Results: None of the studied variables were significantly associated with efficacy of ESI in patients with PHN. PHN that began <11 months before treatment was predictive of a response to ESI at 12 weeks postintervention (positive predictive value, 55%). Patients who reported poor ESI efficacy 2 weeks after the intervention had a 94% chance of still having pain at 12 weeks. Conclusion: For this cohort of patients with PHN being treated with ESI, no demographic characteristics, concurrently used medications, or type of ESI were associated with ESI treatment efficacy at 2 or 12 weeks after the intervention.
Collapse
Affiliation(s)
| | - Christopher S Wie
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - Rhonda S Low
- Division of Preventive, Occupational, and Aerospace Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Nan Zhang
- Biostatistics, Mayo Clinic, Scottsdale, AZ, USA
| | | | | | | |
Collapse
|
5
|
Losartan treatment attenuates the development of neuropathic thermal hyperalgesia induced by peripheral nerve injury in rats. Life Sci 2019; 220:147-155. [DOI: 10.1016/j.lfs.2019.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 12/19/2022]
|
6
|
Abstract
A great need exists for the identification of new effective analgesics to treat sustained pain. However, most preclinical nociceptive assays measure behavioral responses evoked by noxious stimuli (ie, pain-stimulated behavior), which presents a challenge to distinguish between motor impairing and antinociceptive effects of drugs. Here, we demonstrate that chronic constriction injury (CCI) of the sciatic nerve elicits common pain-stimulated responses (ie, mechanical allodynia and thermal hyperalgesia) as well as reduces marble burying/digging behaviors that occur during the early stages of the neuropathy and resolve within 1 week. Although drugs representing distinct classes of analgesics (ie, morphine, valdecoxib, and gabapentin) reversed both CCI-induced and CCI-depressed nociceptive measures, diazepam lacked antinociceptive effects in all assays and the kappa-opioid receptor agonist U69593 reversed pain-stimulated, but not pain-depressed behaviors. In addition, we tested drugs targeting distinct components of the endocannabinoid system, including agonists at cannabinoid receptors type 1 (CB1) and type 2 (CB2), as well as inhibitors of the endocannabinoid-regulating enzymes fatty acid amide hydrolase and monoacylglycerol lipase. Each of these drugs reversed all CCI-induced nociceptive measures, with the exception of the fatty acid amide hydrolase inhibitor that reversed pain-stimulated behaviors, only. These findings support the use of the mouse marble-burying assay as a model of pain-depressed behavior within the first week of sciatic nerve injury to examine candidate analgesics. These data also support existing preclinical research that cannabinoid receptor agonists and inhibitors of endocannabinoid-regulating enzymes merit consideration for the treatment of pain.
Collapse
|
7
|
Poulen G, Perrin FE. Microcebus murinus: A novel promising non-human primate model of spinal cord injury. Neural Regen Res 2018; 13:421-422. [PMID: 29623923 PMCID: PMC5900501 DOI: 10.4103/1673-5374.228721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Gaëtan Poulen
- University of Montpellier, Institut National de la Santé et de la Recherche Médicale Unit 1198; Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Florence Evelyne Perrin
- University of Montpellier, Institut National de la Santé et de la Recherche Médicale Unit 1198, Montpellier, France
| |
Collapse
|
8
|
Spinal mechanisms underlying potentiation of hindpaw responses observed after transient hindpaw ischemia in mice. Sci Rep 2015; 5:11191. [PMID: 26165560 PMCID: PMC4499883 DOI: 10.1038/srep11191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/23/2015] [Indexed: 01/04/2023] Open
Abstract
Transient ischemia produces postischemic tingling sensation. Ischemia also produces nerve conduction block that may modulate spinal neural circuits. In the present study, reduced mechanical thresholds for hindpaw-withdrawal reflex were found in mice after transient hindpaw ischemia, which was produced by a high pressure applied around the hindpaw for 30 min. The reduction in the threshold was blocked by spinal application of LY354740, a specific agonist of group II metabotropic glutamate receptors. Neural activities in the spinal cord and the primary somatosensory cortex (S1) were investigated using activity-dependent changes in endogenous fluorescence derived from mitochondrial flavoproteins. Ischemic treatment induced potentiation of the ipsilateral spinal and contralateral S1 responses to hindpaw stimulation. Both types of potentiation were blocked by spinal application of LY354740. The contralateral S1 responses, abolished by lesioning the ipsilateral dorsal column, reappeared after ischemic treatment, indicating that postischemic tingling sensation reflects a sensory modality shift from tactile sensation to nociception in the spinal cord. Changes in neural responses were investigated during ischemic treatment in the contralateral spinal cord and the ipsilateral S1. Potentiation already appeared during ischemic treatment for 30 min. The present findings suggest that the postischemic potentiation shares spinal mechanisms, at least in part, with neuropathic pain.
Collapse
|
9
|
Schuelert N, Gorodetskaya N, Just S, Doods H, Corradini L. Electrophysiological characterization of spinal neurons in different models of diabetes type 1- and type 2-induced neuropathy in rats. Neuroscience 2015; 291:146-54. [PMID: 25686525 DOI: 10.1016/j.neuroscience.2015.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/15/2015] [Accepted: 02/03/2015] [Indexed: 10/24/2022]
Abstract
Diabetic polyneuropathy (DPN) is a devastating complication of diabetes. The underlying pathogenesis of DPN is still elusive and an effective treatment devoid of side effects presents a challenge. There is evidence that in type-1 and -2 diabetes, metabolic and morphological changes lead to peripheral nerve damage and altered central nociceptive transmission, which may contribute to neuropathic pain symptoms. We characterized the electrophysiological response properties of spinal wide dynamic range (WDR) neurons in three diabetic models. The streptozotocin (STZ) model was used as a drug-induced model of type-1 diabetes, and the BioBreeding/Worcester (BB/Wor) and Zucker diabetic fatty (ZDF) rat models were used for genetic DPN models. Data were compared to the respective control group (BB/Wor diabetic-resistant, Zucker lean (ZL) and saline-injected Wistar rat). Response properties of WDR neurons to mechanical stimulation and spontaneous activity were assessed. We found abnormal response properties of spinal WDR neurons in all diabetic rats but not controls. Profound differences between models were observed. In BB/Wor diabetic rats evoked responses were increased, while in ZDF rats spontaneous activity was increased and in STZ rats mainly after discharges were increased. The abnormal response properties of neurons might indicate differential pathological, diabetes-induced, changes in spinal neuronal transmission. This study shows for the first time that specific electrophysiological response properties are characteristic for certain models of DPN and that these might reflect the diverse and complex symptomatology of DPN in the clinic.
Collapse
Affiliation(s)
- N Schuelert
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany.
| | - N Gorodetskaya
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany
| | - S Just
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany
| | - H Doods
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany
| | - L Corradini
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, 88397 Biberach, Germany
| |
Collapse
|
10
|
Nielson JL, Haefeli J, Salegio EA, Liu AW, Guandique CF, Stück ED, Hawbecker S, Moseanko R, Strand SC, Zdunowski S, Brock JH, Roy RR, Rosenzweig ES, Nout-Lomas YS, Courtine G, Havton LA, Steward O, Reggie Edgerton V, Tuszynski MH, Beattie MS, Bresnahan JC, Ferguson AR. Leveraging biomedical informatics for assessing plasticity and repair in primate spinal cord injury. Brain Res 2014; 1619:124-38. [PMID: 25451131 DOI: 10.1016/j.brainres.2014.10.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 11/18/2022]
Abstract
Recent preclinical advances highlight the therapeutic potential of treatments aimed at boosting regeneration and plasticity of spinal circuitry damaged by spinal cord injury (SCI). With several promising candidates being considered for translation into clinical trials, the SCI community has called for a non-human primate model as a crucial validation step to test efficacy and validity of these therapies prior to human testing. The present paper reviews the previous and ongoing efforts of the California Spinal Cord Consortium (CSCC), a multidisciplinary team of experts from 5 University of California medical and research centers, to develop this crucial translational SCI model. We focus on the growing volumes of high resolution data collected by the CSCC, and our efforts to develop a biomedical informatics framework aimed at leveraging multidimensional data to monitor plasticity and repair targeting recovery of hand and arm function. Although the main focus of many researchers is the restoration of voluntary motor control, we also describe our ongoing efforts to add assessments of sensory function, including pain, vital signs during surgery, and recovery of bladder and bowel function. By pooling our multidimensional data resources and building a unified database infrastructure for this clinically relevant translational model of SCI, we are now in a unique position to test promising therapeutic strategies' efficacy on the entire syndrome of SCI. We review analyses highlighting the intersection between motor, sensory, autonomic and pathological contributions to the overall restoration of function. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Jessica L Nielson
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Jenny Haefeli
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Ernesto A Salegio
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Aiwen W Liu
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Cristian F Guandique
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Ellen D Stück
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Stephanie Hawbecker
- California National Primate Research Center (CNPRC), University of California, Davis, CA (UCD), United States
| | - Rod Moseanko
- California National Primate Research Center (CNPRC), University of California, Davis, CA (UCD), United States
| | - Sarah C Strand
- California National Primate Research Center (CNPRC), University of California, Davis, CA (UCD), United States
| | - Sharon Zdunowski
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA (UCLA), United States
| | - John H Brock
- Center for Neural Repair, Department of Neurosciences, University of California, San Diego, La Jolla, CA (UCSD), United States
| | - Roland R Roy
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA (UCLA), United States
| | - Ephron S Rosenzweig
- Center for Neural Repair, Department of Neurosciences, University of California, San Diego, La Jolla, CA (UCSD), United States
| | - Yvette S Nout-Lomas
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, United States
| | - Gregoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), United States
| | - Leif A Havton
- Reeve-Irvine Research Center (RIRC), University of California, Irvine, CA (UCI), United States; Departments of Anesthesiology & Perioperative Care, Neurology, and Anatomy & Neurobiology, University of California, Irvine, CA, United States
| | - Oswald Steward
- Reeve-Irvine Research Center (RIRC), University of California, Irvine, CA (UCI), United States; Departments of Anatomy & Neurobiology, Neurobiology & Behavior, and Neurosurgery, University of California, Irvine, CA, United States
| | - V Reggie Edgerton
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA (UCLA), United States
| | - Mark H Tuszynski
- Departments of Anesthesiology & Perioperative Care, Neurology, and Anatomy & Neurobiology, University of California, Irvine, CA, United States; Veterans Administration Medical Center, La Jolla, CA, United States
| | - Michael S Beattie
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Jacqueline C Bresnahan
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States
| | - Adam R Ferguson
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA (UCSF), United States.
| |
Collapse
|
11
|
Salmon and human thrombin differentially regulate radicular pain, glial-induced inflammation and spinal neuronal excitability through protease-activated receptor-1. PLoS One 2013; 8:e80006. [PMID: 24278231 PMCID: PMC3835785 DOI: 10.1371/journal.pone.0080006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/07/2013] [Indexed: 11/22/2022] Open
Abstract
Chronic neck pain is a major problem with common causes including disc herniation and spondylosis that compress the spinal nerve roots. Cervical nerve root compression in the rat produces sustained behavioral hypersensitivity, due in part to the early upregulation of pro-inflammatory cytokines, the sustained hyperexcitability of neurons in the spinal cord and degeneration in the injured nerve root. Through its activation of the protease-activated receptor-1 (PAR1), mammalian thrombin can enhance pain and inflammation; yet at lower concentrations it is also capable of transiently attenuating pain which suggests that PAR1 activation rate may affect pain maintenance. Interestingly, salmon-derived fibrin, which contains salmon thrombin, attenuates nerve root-induced pain and inflammation, but the mechanisms of action leading to its analgesia are unknown. This study evaluates the effects of salmon thrombin on nerve root-mediated pain, axonal degeneration in the root, spinal neuronal hyperexcitability and inflammation compared to its human counterpart in the context of their enzymatic capabilities towards coagulation substrates and PAR1. Salmon thrombin significantly reduces behavioral sensitivity, preserves neuronal myelination, reduces macrophage infiltration in the injured nerve root and significantly decreases spinal neuronal hyperexcitability after painful root compression in the rat; whereas human thrombin has no effect. Unlike salmon thrombin, human thrombin upregulates the transcription of IL-1β and TNF-α and the secretion of IL-6 by cortical cultures. Salmon and human thrombins cleave human fibrinogen-derived peptides and form clots with fibrinogen with similar enzymatic activities, but salmon thrombin retains a higher enzymatic activity towards coagulation substrates in the presence of antithrombin III and hirudin compared to human thrombin. Conversely, salmon thrombin activates a PAR1-derived peptide more weakly than human thrombin. These results are the first to demonstrate that salmon thrombin has unique analgesic, neuroprotective and anti-inflammatory capabilities compared to human thrombin and that PAR1 may contribute to these actions.
Collapse
|
12
|
Zhang S, Nicholson KJ, Smith JR, Gilliland TM, Syré PP, Winkelstein BA. The roles of mechanical compression and chemical irritation in regulating spinal neuronal signaling in painful cervical nerve root injury. STAPP CAR CRASH JOURNAL 2013; 57:219-242. [PMID: 24435733 DOI: 10.4271/2013-22-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Both traumatic and slow-onset disc herniation can directly compress and/or chemically irritate cervical nerve roots, and both types of root injury elicit pain in animal models of radiculopathy. This study investigated the relative contributions of mechanical compression and chemical irritation of the nerve root to spinal regulation of neuronal activity using several outcomes. Modifications of two proteins known to regulate neurotransmission in the spinal cord, the neuropeptide calcitonin gene-related peptide (CGRP) and glutamate transporter 1 (GLT-1), were assessed in a rat model after painful cervical nerve root injuries using a mechanical compression, chemical irritation or their combination of injury. Only injuries with compression induced sustained behavioral hypersensitivity (p≤0.05) for two weeks and significant decreases (p<0.037) in CGRP and GLT-1 immunoreactivity to nearly half that of sham levels in the superficial dorsal horn. Because modification of spinal CGRP and GLT-1 is associated with enhanced excitatory signaling in the spinal cord, a second study evaluated the electrophysiological properties of neurons in the superficial and deeper dorsal horn at day 7 after a painful root compression. The evoked firing rate was significantly increased (p=0.045) after compression and only in the deeper lamina. The painful compression also induced a significant (p=0.002) shift in the percentage of neurons in the superficial lamina classified as low- threshold mechanoreceptive (sham 38%; compression 10%) to those classified as wide dynamic range neurons (sham 43%; compression 74%). Together, these studies highlight mechanical compression as a key modulator of spinal neuronal signaling in the context of radicular injury and pain.
Collapse
Affiliation(s)
- Sijia Zhang
- Department of Bioengineering, University of Pennsylvania
| | | | - Jenell R Smith
- Department of Bioengineering, University of Pennsylvania
| | | | - Peter P Syré
- Department of Neurosurgery, University of Pennsylvania
| | - Beth A Winkelstein
- Departments of Bioengineering and Neurosurgery, University of Pennsylvania
| |
Collapse
|
13
|
Carozzi VA, Renn CL, Bardini M, Fazio G, Chiorazzi A, Meregalli C, Oggioni N, Shanks K, Quartu M, Serra MP, Sala B, Cavaletti G, Dorsey SG. Bortezomib-induced painful peripheral neuropathy: an electrophysiological, behavioral, morphological and mechanistic study in the mouse. PLoS One 2013; 8:e72995. [PMID: 24069168 PMCID: PMC3772181 DOI: 10.1371/journal.pone.0072995] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/23/2013] [Indexed: 11/18/2022] Open
Abstract
Bortezomib is the first proteasome inhibitor with significant antineoplastic activity for the treatment of relapsed/refractory multiple myeloma as well as other hematological and solid neoplasms. Peripheral neurological complications manifesting with paresthesias, burning sensations, dysesthesias, numbness, sensory loss, reduced proprioception and vibratory sensitivity are among the major limiting side effects associated with bortezomib therapy. Although bortezomib-induced painful peripheral neuropathy is clinically easy to diagnose and reliable models are available, its pathophysiology remains partly unclear. In this study we used well-characterized immune-competent and immune-compromised mouse models of bortezomib-induced painful peripheral neuropathy. To characterize the drug-induced pathological changes in the peripheral nervous system, we examined the involvement of spinal cord neuronal function in the development of neuropathic pain and investigated the relevance of the immune response in painful peripheral neuropathy induced by bortezomib. We found that bortezomib treatment induced morphological changes in the spinal cord, dorsal roots, dorsal root ganglia (DRG) and peripheral nerves. Neurophysiological abnormalities and specific functional alterations in Aδ and C fibers were also observed in peripheral nerve fibers. Mice developed mechanical allodynia and functional abnormalities of wide dynamic range neurons in the dorsal horn of spinal cord. Bortezomib induced increased expression of the neuronal stress marker activating transcription factor-3 in most DRG. Moreover, the immunodeficient animals treated with bortezomib developed a painful peripheral neuropathy with the same features observed in the immunocompetent mice. In conclusion, this study extends the knowledge of the sites of damage induced in the nervous system by bortezomib administration. Moreover, a selective functional vulnerability of peripheral nerve fiber subpopulations was found as well as a change in the electrical activity of wide dynamic range neurons of dorsal horn of spinal cord. Finally, the immune response is not a key factor in the development of morphological and functional damage induced by bortezomib in the peripheral nervous system.
Collapse
Affiliation(s)
- Valentina A. Carozzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
- * E-mail: (VAC)
| | - Cynthia L. Renn
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Michela Bardini
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Grazia Fazio
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Alessia Chiorazzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Cristina Meregalli
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Norberto Oggioni
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Kathleen Shanks
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Marina Quartu
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Maria Pina Serra
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Barbara Sala
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Guido Cavaletti
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Susan G. Dorsey
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|
14
|
Somers DL, Clemente RF. The neurophysiological basis of peripheral electrical nerve stimulation for the treatment of causalgia. PHYSICAL THERAPY REVIEWS 2013. [DOI: 10.1179/ptr.1996.1.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
15
|
Sikandar S, Ronga I, Iannetti GD, Dickenson AH. Neural coding of nociceptive stimuli—from rat spinal neurones to human perception. Pain 2013; 154:1263-73. [DOI: 10.1016/j.pain.2013.03.041] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 03/21/2013] [Accepted: 03/29/2013] [Indexed: 11/26/2022]
|
16
|
Suzuki I, Tsuboi Y, Shinoda M, Shibuta K, Honda K, Katagiri A, Kiyomoto M, Sessle BJ, Matsuura S, Ohara K, Urata K, Iwata K. Involvement of ERK phosphorylation of trigeminal spinal subnucleus caudalis neurons in thermal hypersensitivity in rats with infraorbital nerve injury. PLoS One 2013; 8:e57278. [PMID: 23451198 PMCID: PMC3579857 DOI: 10.1371/journal.pone.0057278] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 01/23/2013] [Indexed: 01/19/2023] Open
Abstract
To evaluate the involvement of the mitogen-activated protein kinase (MAPK) cascade in orofacial neuropathic pain mechanisms, this study assessed nocifensive behavior evoked by mechanical or thermal stimulation of the whisker pad skin, phosphorylation of extracellular signal-regulated kinase (ERK) in trigeminal spinal subnucleus caudalis (Vc) neurons, and Vc neuronal responses to mechanical or thermal stimulation of the whisker pad skin in rats with the chronic constriction nerve injury of the infraorbital nerve (ION-CCI). The mechanical and thermal nocifensive behavior was significantly enhanced on the side ipsilateral to the ION-CCI compared to the contralateral whisker pad or sham rats. ION-CCI rats had an increased number of phosphorylated ERK immunoreactive (pERK-IR) cells which also manifested NeuN-IR but not GFAP-IR and Iba1-IR, and were significantly more in ION-CCI rats compared with sham rats following noxious but not non-noxious mechanical stimulation. After intrathecal administration of the MEK1 inhibitor PD98059 in ION-CCI rats, the number of pERK-IR cells after noxious stimulation and the enhanced thermal nocifensive behavior but not the mechanical nocifensive behavior were significantly reduced in ION-CCI rats. The enhanced background activities, afterdischarges and responses of wide dynamic range neurons to noxious mechanical and thermal stimulation in ION-CCI rats were significantly depressed following i.t. administration of PD98059, whereas responses to non-noxious mechanical and thermal stimulation were not altered. The present findings suggest that pERK-IR neurons in the Vc play a pivotal role in the development of thermal hypersensitivity in the face following trigeminal nerve injury.
Collapse
Affiliation(s)
- Ikuko Suzuki
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Yoshiyuki Tsuboi
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Kazuo Shibuta
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Kuniya Honda
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Ayano Katagiri
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Masaaki Kiyomoto
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Barry J. Sessle
- Department of Oral Physiology, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Shingo Matsuura
- Department of Endodontics, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Kinuyo Ohara
- Department of Endodontics, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Kentaro Urata
- Department of Prosthodontics, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Chiyoda-ku Tokyo, Japan
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
- Division of Applied System Neuroscience Advanced Medical Research Center, Nihon University Graduate School of Medical Science, Tokyo, Japan
| |
Collapse
|
17
|
Guo N, Gu X, Zhao J, Zhao G, Jin M, Zou H, Zhang Y, Zhao Z, Jin GJ, Yu L. Maxillary nerve compression in cynomolgus monkey Macaca fascicularis: altered somatic sensation and peripheral nerve firing. BMC Neurosci 2012; 13:150. [PMID: 23234480 PMCID: PMC3554490 DOI: 10.1186/1471-2202-13-150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 11/29/2012] [Indexed: 01/01/2023] Open
Abstract
Background Trigeminal nerve is a major source of the sensory input of the face, and trigeminal neuropathology models have been reported in rodents with injury to branches of the maxillary or mandibular division of the trigeminal nerve. Non-human primates are neuroanatomically more closely related to human than rodents; however, nerve injury studies in non-human primates are limited. Results We describe here a nerve injury model of maxillary nerve compression (MNC) in the cynomolgus macaque monkey, Macaca fascicularis, and the initial characterization of the consequences of damage to this trigeminal nerve branch. The nerve injury from the compression appeared to be mild, as we did not observe overt changes in home-cage behavior in the monkeys. When mechanical stimulation was applied to the facial area, monkeys with MNC displayed increased mechanical sensitivity, as the avoidance response scores were lower than those from the control animals. Such a change in mechanical sensitivity appeared to be somewhat bilateral, as the contralateral side also showed increased mechanical sensitivity, although the change on the ipsilateral side was more robust. Multiple-unit recording of the maxillary nerve showed a general pattern of increasing responsiveness to escalating force in mechanical stimulation on the contralateral side. Ipsilateral side of the maxillary nerve showed a lack of responsiveness to escalating force in mechanical stimulation, possibly reflecting a maximum stimulation threshold effect from sensitized nerve due to MNC injury. Conclusions These results suggest that MNC may produce increased sensitivity of the ipsilateral maxillary nerve, and that this model may serve as a non-human primate model to evaluate the effect of injury to trigeminal nerve branches.
Collapse
Affiliation(s)
- Ning Guo
- Shanghai University of Traditional Chinese Medicine, and ShanghaiBio Corporation, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Engholm G, Leffler AS. Influence of pain reduction by transcutaneous electrical nerve stimulation (TENS) on somatosensory functions in patients with painful traumatic peripheral partial nerve injury. Eur J Pain 2012; 14:918-23. [DOI: 10.1016/j.ejpain.2010.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 02/08/2010] [Accepted: 02/17/2010] [Indexed: 10/19/2022]
|
19
|
Carlton SM, Rees H, Tsuruoka M, Willis WD. Memantine attenuates responses of spinothalamic tract cells to cutaneous stimulation in neuropathic monkeys. Eur J Pain 2012; 2:229-38. [PMID: 15102383 DOI: 10.1016/s1090-3801(98)90019-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/1998] [Accepted: 05/06/1998] [Indexed: 11/23/2022]
Abstract
Several lines of evidence indicate that N-methyl-D-aspartate (NMDA) receptors play an important role in nociception in general and in pathological pain in particular. It has been previously demonstrated in behavioral studies that NMDA receptor antagonists attenuate pathological pain in humans and nociceptive behaviors in animals. In the present study, we investigated the effect of the NMDA receptor antagonist memantine (MEM) on the responses of spinothalamic tract (STT) cells in normal and neuropathic monkeys. Memantine was delivered into the spinal cord through a microdialysis fiber acutely implanted into the dorsal horn. Responses of STT cells to peripheral stimulation within their receptive fields were recorded before and after MEM infusion. In normal animals (n = 7), 10 mm MEM did not affect STT cell (n = 7) baseline activity or responses to mechanical stimuli (brush, press or pinch). In neuropathic animals (n = 6), 1.0, 3.0, 10.0 and 100 mm MEM did not affect baseline activity of STT cells (n = 7); however, in a dose-dependent fashion, it significantly reduced responses of these cells to all cutaneous stimuli. The data suggest that MEM can have a direct effect on STT cells, blocking NMDA receptors known to be present on this cell population and, furthermore, may be a therapeutic agent for chronic pain.
Collapse
Affiliation(s)
- S M Carlton
- Department of Anatomy and Neurosciences, Marine Biomedical Institute, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | |
Collapse
|
20
|
Leffler AS, Hansson P. Painful traumatic peripheral partial nerve injury-sensory dysfunction profiles comparing outcomes of bedside examination and quantitative sensory testing. Eur J Pain 2012; 12:397-402. [DOI: 10.1016/j.ejpain.2007.08.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 08/17/2007] [Accepted: 08/30/2007] [Indexed: 11/16/2022]
|
21
|
Jergova S, Hentall ID, Gajavelli S, Varghese MS, Sagen J. Intraspinal transplantation of GABAergic neural progenitors attenuates neuropathic pain in rats: a pharmacologic and neurophysiological evaluation. Exp Neurol 2011; 234:39-49. [PMID: 22193109 DOI: 10.1016/j.expneurol.2011.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/27/2011] [Accepted: 12/05/2011] [Indexed: 02/03/2023]
Abstract
Dysfunctional γ-aminobutyric acid (GABA)-ergic inhibitory neurotransmission is hypothesized to underlie chronic neuropathic pain. Intraspinal transplantation of GABAergic neural progenitor cells (NPCs) may reduce neuropathic pain by restoring dorsal horn inhibition. Rat NPCs pre-differentiated to a GABAergic phenotype were transplanted into the dorsal horn of rats with unilateral chronic constriction injury (CCI) of the sciatic nerve. GABA signaling in antinociceptive effects of NPC grafts was tested with the GABA(A) receptor antagonist bicuculline (BIC), GABA(B) receptor antagonist CGP35348 (CGP) and GABA reuptake inhibitor SKF 89976A (SKF). NPC-treated animals showed decreased hyperalgesia and allodynia 1-3week post-transplantation; vehicle-injected CCI rats continued displaying pain behaviors. Intrathecal application of BIC or CGP attenuated the antinociceptive effects of the NPC transplants while SKF injection induced analgesia in control rats. Electrophysiological recordings in NPC treated rats showed reduced responses of wide dynamic range (WDR) neurons to peripheral stimulation compared to controls. A spinal application of BIC or CGP increased wind-up response and post-discharges of WDR neurons in NPC treated animals. Results suggest that transplantation of GABAergic NPCs attenuate pain behaviors and reduce exaggerated dorsal horn neuronal firing induced by CCI. The effects of GABA receptor inhibitors suggest participation of continuously released GABA in the grafted animals.
Collapse
Affiliation(s)
- Stanislava Jergova
- University of Miami, Miller School of Medicine, Miami Project to Cure Paralysis, 1095 NW 14 Terrace, Miami, Florida 33136, USA.
| | | | | | | | | |
Collapse
|
22
|
Renn CL, Leitch CC, Lessans S, Rhee P, McGuire WC, Smith BA, Traub RJ, Dorsey SG. Brain-derived neurotrophic factor modulates antiretroviral-induced mechanical allodynia in the mouse. J Neurosci Res 2011; 89:1551-65. [PMID: 21647939 DOI: 10.1002/jnr.22685] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/06/2011] [Accepted: 04/12/2011] [Indexed: 01/12/2023]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are key components of HIV/AIDS treatment to reduce viral load. However, these drugs can induce chronic neuropathic pain, leading to increased morbidity in HIV patients. This study examines the role of brain-derived neurotrophic factor (BDNF) in the spinal dorsal horn (SDH) in development of mechanical allodynia in male C57BL/6J mice treated with the NRTI stavudine (d4T). After d4T administration, mice developed increased neuronal activity and BDNF expression in the SDH and hind paw mechanical allodynia that was exacerbated by intrathecal BDNF administration. Intrathecal BDNF alone also increased neuronal activity and caused mechanical allodynia. Because excess BDNF amplified d4T-induced mechanical allodynia and neuronal activity, the impact of decreasing BDNF in the SDH was investigated. After d4T, BDNF heterozygous mice were less allodynic than wild-type littermates, which was negated by intrathecal BDNF administration. Finally, pretreatment with intrathecal trkB-Fc chimera prior to d4T or administration of the tyrosine kinase inhibitor K252a 3 days after d4T blocked BDNF-mediated signaling, significantly attenuated the development of mechanical allodynia (trkB-Fc), and decreased neuronal activity (trkB-Fc and K252a). Taken together, these findings provide evidence that BDNF in the SDH contributes to the development of NRTI-induced painful peripheral neuropathy and may represent a new therapeutic opportunity.
Collapse
Affiliation(s)
- Cynthia L Renn
- Department of Organizational Systems and Adult Health, School of Nursing, University of Maryland, Baltimore, Maryland 21201-1579, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
We tested a hypothesis that the spinal plasticity induced within a few hours after nerve injury may produce changes in cortical activities and an initial phase of neuropathic pain. Somatosensory cortical responses elicited by vibratory stimulation were visualized by transcranial flavoprotein fluorescence imaging in mice. These responses were reduced immediately after cutting the sensory nerves. However, the remaining cortical responses mediated by nearby nerves were potentiated within a few hours after nerve cutting. Nerve injury induces neuropathic pain. In the present study, mice exhibited tactile allodynia 1-2 weeks after nerve injury. Lesioning of the ipsilateral dorsal column, mediating tactile cortical responses, abolished somatic cortical responses to tactile stimuli. However, nontactile cortical responses appeared in response to the same tactile stimuli within a few hours after nerve injury, indicating that tactile allodynia was acutely initiated. We investigated the trigger mechanisms underlying the cortical changes. Endogenous glial cell line-derived neurotrophic factor (GDNF), found in the Meissner corpuscles, induced basal firing ∼0.1 Hz or less in its Aβ tactile afferents, and disruption of the basal firing triggered the potentiation of nontactile cortical responses. Application of 10 nm LY341495 [(2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl) propanoic acid], a specific antagonist of group II metabotropic glutamate receptors (mGluRs), on to the surface of the spinal cord also induced the potentiation of nontactile cortical responses. Together, it is suggested that low-frequency afferent firing produced by GDNF in touch-sensitive nerve fibers continuously activated spinal group II mGluRs and that failure of this activation triggered tactile allodynia.
Collapse
|
24
|
Ushida T, Fukumoto M, Binti C, Ikemoto T, Taniguchi S, Ikeuchi M, Nishihara M, Tani T. Alterations of contralateral thalamic perfusion in neuropathic pain. Open Neuroimag J 2010; 4:182-6. [PMID: 21347202 PMCID: PMC3043277 DOI: 10.2174/1874440001004010182] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 12/15/2009] [Accepted: 02/12/2010] [Indexed: 11/30/2022] Open
Abstract
Contralateral thalamus, the place of termination of spinothalamic tract, is affected in patients with pain. We employed single photon emission computed tomography (SPECT) to evaluate the thalamic perfusion in patients with spontaneous neuropathic pain. Ten patients with complex regional pain syndrome (CRPS) and eleven radiculopathiy patients were enrolled in this study. Regional cerebral blood flow of thalamus was assessed bilaterally by iodine-123-labelled iodoamphetamine SPECT. To standardize the inter-patient data, we set a contralateral thalamic uptake index (CTUI) for assessing thalamic asymmetry. In one study, we found elevation of CTUI in patients with symptoms of neuropathic pain for less than 12 month, whereas no change was observed in the case of a longer lasting disease. An another study demonstrated decrease of CTUI after pain treatment, even though it was unrelated to the pain intensity prior to treatment. Our SPECT study revealed that neuropathic pain altered thalamic neuronal activity. CTUIs were increased in early stage of the disease but decreased as the disease progressed to the chronic stage. These results suggest that CTUI can be used to improve management of neuropathic pain for proper evaluation of spontaneous pain.
Collapse
Affiliation(s)
- Takahiro Ushida
- Department of Orthopaedic Surgery Kochi Medical School, Kochi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Quinn KP, Dong L, Golder FJ, Winkelstein BA. Neuronal hyperexcitability in the dorsal horn after painful facet joint injury. Pain 2010; 151:414-421. [PMID: 20739124 DOI: 10.1016/j.pain.2010.07.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 06/23/2010] [Accepted: 07/31/2010] [Indexed: 12/24/2022]
Abstract
Excessive cervical facet capsular ligament stretch has been implicated as a cause of whiplash-associated disorders following rear-end impacts, but the pathophysiological mechanisms that produce chronic pain in these cases remain unclear. Using a rat model of C6-C7 cervical facet joint capsule stretch that produces sustained mechanical hyperalgesia, the presence of neuronal hyperexcitability was characterized 7 days after joint loading. Extracellular recordings of spinal dorsal horn neuronal activity between C6 and C8 (117 neurons) were obtained from anesthetized rats, with both painful and non-painful behavioral outcomes established by the magnitude of capsule stretch. The frequency of neuronal firing during noxious pinch (p<0.0182) and von Frey filaments applications (4-26g) to the forepaw was increased (p<0.0156) in the painful group compared to the non-painful and sham groups. In addition, the incidence and frequency of spontaneous and after discharge firing were greater in the painful group (p<0.0307) relative to sham. The proportion of cells in the deep laminae that responded as wide dynamic range neurons also was increased in the painful group relative to non-painful or sham groups (p<0.0348). These findings suggest that excessive facet capsule stretch, while not producing visible tearing, can produce functional plasticity of dorsal horn neuronal activity. The increase in neuronal firing across a range of stimulus magnitudes observed at day 7 post-injury provides the first direct evidence of neuronal modulation in the spinal cord following facet joint loading, and suggests that facet-mediated chronic pain following whiplash injury is driven, at least in part, by central sensitization.
Collapse
Affiliation(s)
- Kyle P Quinn
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA Department of Clinical Studies - Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
26
|
Nakagawa K, Takeda M, Tsuboi Y, Kondo M, Kitagawa J, Matsumoto S, Kobayashi A, Sessle BJ, Shinoda M, Iwata K. Alteration of primary afferent activity following inferior alveolar nerve transection in rats. Mol Pain 2010; 6:9. [PMID: 20122287 PMCID: PMC2829527 DOI: 10.1186/1744-8069-6-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/03/2010] [Indexed: 01/17/2023] Open
Abstract
Background In order to evaluate the neural mechanisms underlying the abnormal facial pain that may develop following regeneration of the injured inferior alveolar nerve (IAN), the properties of the IAN innervated in the mental region were analyzed. Results Fluorogold (FG) injection into the mental region 14 days after IAN transection showed massive labeling of trigeminal ganglion (TG). The escape threshold to mechanical stimulation of the mental skin was significantly lower (i.e. mechanical allodynia) at 11-14 days after IAN transection than before surgery. The background activity, mechanically evoked responses and afterdischarges of IAN Aδ-fibers were significantly higher in IAN-transected rats than naive. The small/medium diameter TG neurons showed an increase in both tetrodotoxin (TTX)-resistant (TTX-R) and -sensitive (TTX-S) sodium currents (INa) and decrease in total potassium current, transient current (IA) and sustained current (IK) in IAN-transected rats. The amplitude, overshoot amplitude and number of action potentials evoked by the depolarizing pulses after 1 μM TTX administration in TG neurons were significantly higher, whereas the threshold current to elicit spikes was smaller in IAN-transected rats than naive. Resting membrane potential was significantly smaller in IAN-transected rats than that of naive. Conclusions These data suggest that the increase in both TTX-S INa and TTX-R INa and the decrease in IA and Ik in small/medium TG neurons in IAN-transected rats are involved in the activation of spike generation, resulting in hyperexcitability of Aδ-IAN fibers innervating the mental region after IAN transection.
Collapse
Affiliation(s)
- Kazuharu Nakagawa
- Department of Dysphagia Rehabilitation, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kupers R, Schneider FCG, Christensen R, Naert A, Husted H, Paulson OB, Kehlet H. No Evidence for Generalized Increased Postoperative Responsiveness to Pain: A Combined Behavioral and Serial Functional Magnetic Resonance Imaging Study. Anesth Analg 2009; 109:600-6. [DOI: 10.1213/ane.0b013e3181ac1866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
28
|
|
29
|
Hulsebosch CE, Hains BC, Crown ED, Carlton SM. Mechanisms of chronic central neuropathic pain after spinal cord injury. ACTA ACUST UNITED AC 2008; 60:202-13. [PMID: 19154757 DOI: 10.1016/j.brainresrev.2008.12.010] [Citation(s) in RCA: 232] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 12/25/2022]
Abstract
Not all spinal contusions result in mechanical allodynia, in which non-noxious stimuli become noxious. The studies presented use the NYU impactor at 12.5 mm drop or the Infinite Horizons Impactor (150 kdyn, 1 s dwell) devices to model spinal cord injury (SCI). Both of these devices and injury parameters, if done correctly, will result in animals with above level (forelimb), at level (trunk) and below level (hindlimb) mechanical allodynia that model the changes in evoked somatosensation experienced by the majority of people with SCI. The sections are as follows: 1) Mechanisms of remote microglial activation and pain signaling in "below-level" central pain 2) Intracellular signaling mechanisms in central sensitization in "at-level" pain 3) Peripheral sensitization contributes to "above level" injury pain following spinal cord injury and 4) Role of reactive oxygen species in central sensitization in regional neuropathic pain following SCI. To summarize, differential regional mechanisms contribute to the regional chronic pain states. We propose the importance of understanding the mechanisms in the differential regional pain syndromes after SCI in the chronic condition. Targeting regional mechanisms will be of enormous benefit to the SCI population that suffer chronic pain, and will contribute to better treatment strategies for other chronic pain syndromes.
Collapse
Affiliation(s)
- Claire E Hulsebosch
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA.
| | | | | | | |
Collapse
|
30
|
Behavioral and electrophysiological studies in rats with cisplatin-induced chemoneuropathy. Brain Res 2008; 1230:91-8. [PMID: 18657527 DOI: 10.1016/j.brainres.2008.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 07/02/2008] [Accepted: 07/07/2008] [Indexed: 11/20/2022]
Abstract
Neuropathy is the chief dose-limiting side effect associated with the major classes of frontline cancer therapy drugs. Here the changes in behavioral responses of rats to cutaneous mechanical and thermal stimuli occurring following treatment with cisplatin and the changes in spinal neurophysiology accompanying the development of chemotherapy-induced hyperalgesia were explored. Systemic treatment with cisplatin induced changes in both mechanical and thermal cutaneous sensory withdrawal thresholds of Sprague-Dawley rats. High doses of chemotherapy produced hypoalgesia whereas lower doses produced hyperalgesia. Follow-up neurophysiological studies in rats with chemotherapy-induced hyperalgesia revealed that deep spinal lamina wide dynamic range neurons had significantly higher spontaneous activity and longer afterdischarges to noxious mechanical stimuli than wide dynamic range neurons in control rats; cisplatin administration was also associated with longer afterdischarges and abnormal wind-up to transcutaneous electrical stimuli. The hyperexcitability observed during cisplatin-induced hyperalgesia is very similar to that observed in rats with hyperalgesia produced following treatment with other very diverse types of chemotherapeutic agents and similar to that observed following specific types of direct nerve injury.
Collapse
|
31
|
Saito K, Hitomi S, Suzuki I, Masuda Y, Kitagawa J, Tsuboi Y, Kondo M, Sessle BJ, Iwata K. Modulation of Trigeminal Spinal Subnucleus Caudalis Neuronal Activity Following Regeneration of Transected Inferior Alveolar Nerve in Rats. J Neurophysiol 2008; 99:2251-63. [DOI: 10.1152/jn.00794.2007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Modulation of trigeminal spinal subnucleus caudalis neuronal activity following regeneration of transected inferior alveolar nerve in rats. To clarify the neuronal mechanisms of abnormal pain in the face innervated by the regenerated inferior alveolar nerve (IAN), nocifensive behavior, trigeminal ganglion neuronal labeling following Fluorogold (FG) injection into the mental skin, and trigeminal spinal subnucleus caudalis (Vc) neuronal properties were examined in rats with IAN transection. The mechanical escape threshold was significantly higher at 3 days and lower at 14 days after IAN transection, whereas head withdrawal latency to heat was significantly longer at 3, 14, and 60 days after IAN transection. The number of FG-labeled ganglion neurons was significantly reduced at 3 days after IAN transection but increased at 14 and 60 days. The number of wide dynamic range (WDR) neurons with background (BG) activity was significantly higher at 14 and 60 days after IAN transection compared with naïve rats, and the number of WDR and low-threshold mechanoreceptive (LTM) neurons with irregularly bursting BG activity was increased at these two time points. Mechanically evoked responses were significantly larger in WDR and LTM neurons 14 days after IAN transection compared with naïve rats. Heat- and cold-evoked responses in WDR neurons were significantly lower at 14 days after transection compared with naïve rats. Mechanoreceptive fields were also significantly larger in WDR and LTM neurons at 14 and 60 days after IAN transection. These findings suggest that these alterations may be involved in the development of mechanical allodynia in the cutaneous region innervated by the regenerated IAN.
Collapse
|
32
|
Carlson JD, Maire JJ, Martenson ME, Heinricher MM. Sensitization of pain-modulating neurons in the rostral ventromedial medulla after peripheral nerve injury. J Neurosci 2007; 27:13222-31. [PMID: 18045916 PMCID: PMC6673414 DOI: 10.1523/jneurosci.3715-07.2007] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 10/01/2007] [Accepted: 10/24/2007] [Indexed: 12/13/2022] Open
Abstract
Nerve injury can lead to mechanical hypersensitivity in both humans and animal models, such that innocuous touch produces pain. Recent functional studies have demonstrated a critical role for descending pain-facilitating influences from the rostral ventromedial medulla (RVM) in neuropathic pain, but the underlying mechanisms and properties of the relevant neurons within the RVM are essentially unknown. We therefore characterized mechanical responsiveness of physiologically characterized neurons in the RVM after spinal nerve ligation, a model of neuropathic pain that produces robust mechanical hyperalgesia and allodynia. RVM neurons were studied 7-14 d after spinal nerve ligation, and classified as "on-cells," "off-cells," or "neutral cells" using standard criteria of changes in firing associated with heat-evoked reflexes. On-cells are known to promote nociception, and off-cells to suppress nociception, whereas the role of neutral cells in pain modulation remains an open question. Neuronal and behavioral responses to innocuous and noxious mechanical stimulation were tested using calibrated von Frey filaments (4-100 g) applied to the hindpaws ipsilateral and contralateral to the injury, and in sham-operated and unoperated control animals. On- and off-cells recorded in nerve-injured animals exhibited novel responses to innocuous mechanical stimulation, and enhanced responses to noxious mechanical stimulation. Neuronal hypersensitivity in the RVM was correlated with behavioral hypersensitivity. Neutral cells remained unresponsive to cutaneous stimulation after nerve injury. These data demonstrate that both on- and off-cells in the RVM are sensitized to innocuous and noxious mechanical stimuli after nerve injury. This sensitization likely contributes to allodynia and hyperalgesia of neuropathic pain states.
Collapse
Affiliation(s)
- Jonathan D. Carlson
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| | - Jennifer J. Maire
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| | - Melissa E. Martenson
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| | - Mary M. Heinricher
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
33
|
Ji G, Zhou S, Kochukov MY, Westlund KN, Carlton SM. Plasticity in intact A delta- and C-fibers contributes to cold hypersensitivity in neuropathic rats. Neuroscience 2007; 150:182-93. [PMID: 17945425 DOI: 10.1016/j.neuroscience.2007.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 09/10/2007] [Accepted: 09/12/2007] [Indexed: 10/22/2022]
Abstract
Cold hypersensitivity is a common sensory abnormality accompanying peripheral neuropathies and is difficult to treat. Progress has been made in understanding peripheral mechanisms underlying neuropathic pain but little is known concerning peripheral mechanisms of cold hypersensitivity. The aim of this study was to analyze the contribution of uninjured primary afferents to the cold hypersensitivity that develops in neuropathic rats. Rats with a lumbar 5 (L5) and L6 spinal nerve ligation (SNL, Chung model) but not sham, developed mechanical allodynia, evidenced by decreased paw withdrawal thresholds and increased magnitude of response to von Frey stimulation. Cold hypersensitivity also developed in SNL but not sham rats, evidenced by enhanced nociceptive behaviors induced by placement on a cold plate (6 degrees C) or application of icilin (a transient receptor potential M8 (TRPM8)/transient receptor potential A1 (TRPA1) receptor agonist) to nerve-injured hind paws. Single fiber recordings demonstrated that the mean conduction velocities of intact L4 cutaneous A delta- and C-fibers were not different between naive and SNL rats; however, mechanical thresholds of the A delta- but not the C-fibers were significantly decreased in SNL compared with naive. There was a higher prevalence of C-mechanoheat-cold (CMHC) fibers in SNL compared with naive, but the overall percentage of cold-sensitive C-fibers was not significantly increased compared with naive. This was in contrast to the numerous changes in A delta-fibers: the percentage of L4 cold sensitive A delta-, but not C-fibers, was significantly increased, the percentage of L4 icilin-sensitive A delta-, but not C-fibers, was significantly increased, the icilin-induced activity of L4 A delta-, but not C-fibers, was significantly increased. Icilin-induced activity was blocked by the TRPA1 antagonist Ruthenium Red. The results indicate plasticity in both A delta- and C-uninjured fibers, but A delta fibers appear to provide a major contribution to cold hypersensitivity in neuropathic rats.
Collapse
Affiliation(s)
- G Ji
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Marine Biomedical Institute, 301 University Boulevard, Galveston, TX 77555-1069, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
Pain research has uncovered important neuronal mechanisms that underlie clinically relevant pain states such as inflammatory and neuropathic pain. Importantly, both the peripheral and the central nociceptive system contribute significantly to the generation of pain upon inflammation and nerve injury. Peripheral nociceptors are sensitized during inflammation, and peripheral nerve fibres develop ectopic discharges upon nerve injury or disease. As a consequence a complex neuronal response is evoked in the spinal cord where neurons become hyperexcitable, and a new balance is set between excitation and inhibition. The spinal processes are significantly influenced by brain stem circuits that inhibit or facilitate spinal nociceptive processing. Numerous mechanisms are involved in peripheral and central nociceptive processes including rapid functional changes of signalling and long-term regulatory changes such as up-regulation of mediator/receptor systems. Conscious pain is generated by thalamocortical networks that produce both sensory discriminative and affective components of the pain response.
Collapse
Affiliation(s)
- H G Schaible
- Institut für Physiologie/Neurophysiologie, Teichgraben 8, 07740 Jena, Germany.
| |
Collapse
|
35
|
Kitagawa J, Takeda M, Suzuki I, Kadoi J, Tsuboi Y, Honda K, Matsumoto S, Nakagawa H, Tanabe A, Iwata K. Mechanisms involved in modulation of trigeminal primary afferent activity in rats with peripheral mononeuropathy. Eur J Neurosci 2006; 24:1976-86. [PMID: 17040479 DOI: 10.1111/j.1460-9568.2006.05065.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In order to clarify the mechanisms underlying the changes in primary afferent neurons in trigeminal neuropathic pain, a chronic constriction nerve injury model of the infraorbital nerve (ION-CCI) was developed in rats. Mechanical allodynia was observed at 3 days after ION-CCI and lasted more than 14 days. Single-unit activities were recorded from the ION of anesthetized rats. C-, Abeta- and Adelta-units were identified on the basis of their conduction velocity. Adelta-units were frequently encountered at a later period after ION-CCI. The highest Adelta-spontaneous activity was recorded at 3 days after ION-CCI and progressively decreased after that, but spontaneous activity was still higher at 14 days after ION-CCI than that of naïve rats. Mechanical-evoked responses of Adelta-units were also highest at 3 days after ION-CCI and then gradually decreased. In consideration of these data, patch-clamp recordings were performed on medium to large size neurons of the dissociated trigeminal ganglion (TRG). Patch-clamp recordings revealed that the IK (sustained) and IA (transient) in rats with ION-CCI were significantly smaller than those of naïve rats, and correlated with an increase in duration of repolarization phase and a decrease in duration of depolarization phase, respectively. The hyperpolarization-activated current (Ih) was significantly larger in TRG neurons of rats with ION-CCI as compared with those of naïve rats. The present results suggest that Ih, IK and IA in Adelta-afferent neurons in TRG are significantly involved in the changes in afferent spontaneous activity and mechanically evoked activity that accompany mechanical allodynia produced by trigeminal nerve injury.
Collapse
Affiliation(s)
- Junichi Kitagawa
- Department of Physiology, School of Dentistry, Nihon University, 1-8-13 Kandasurugadai, Chiyoda-ku Tokyo, 101-8310, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liu C, Walker JM. Effects of a cannabinoid agonist on spinal nociceptive neurons in a rodent model of neuropathic pain. J Neurophysiol 2006; 96:2984-94. [PMID: 16943316 DOI: 10.1152/jn.00498.2006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of the synthetic cannabinoid WIN 55,212-2 on heat-evoked firing of spinal wide dynamic range (WDR) neurons were examined in a rodent model of neuropathic pain. Fifty-eight WDR neurons (1 cell/animal) were recorded from the ipsilateral spinal dorsal horns of rats with chronic constriction injury (CCI) and sham-operated controls. Relative to sham-operated controls, neurons recorded in CCI rats showed elevations in spontaneous firing, noxious heat-evoked responses, and afterdischarge firing as well as increases in receptive field size. WIN 55,212-2 (0.0625, 0.125, and 0.25 mg/kg, intravenous) dose-dependently suppressed heat-evoked activity and decreased the receptive field areas of dorsal horn WDR neurons in both nerve injured and control rats with a greater inhibition in CCI rats. At the dose of 0.125 mg/kg iv, WIN 55,212-2 reversed the hyperalgesia produced by nerve injury. The effect of intravenous administration of WIN 55,212-2 appears to be centrally mediated because administration of the drug directly to the ligated nerve did not suppress the heat-evoked neuronal activity in CCI rats. Pretreatment with the cannabinoid CB(1) receptor antagonists SR141716A or AM251, but not the CB(2) antagonist SR144528, blocked the effects. These results provide a neural basis for reports of potent suppression by cannabinoids of the abnormal sensory responses that result from nerve injury.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Benzoxazines
- Camphanes/pharmacology
- Cannabinoid Receptor Agonists
- Cannabinoid Receptor Antagonists
- Dose-Response Relationship, Drug
- Electrophysiology
- Hot Temperature
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Hyperalgesia/psychology
- In Vitro Techniques
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Neurons/drug effects
- Nociceptors/drug effects
- Pain/drug therapy
- Pain/etiology
- Pain/physiopathology
- Peripheral Nervous System Diseases/complications
- Peripheral Nervous System Diseases/physiopathology
- Piperidines/pharmacology
- Posterior Horn Cells/physiology
- Pyrazoles/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/drug effects
- Rimonabant
- Spinal Cord/cytology
- Spinal Cord/drug effects
- Spinal Cord/physiopathology
Collapse
Affiliation(s)
- Cheng Liu
- Department of Psychological and Brain Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405-7007, USA.
| | | |
Collapse
|
37
|
Noguchi K. Chapter 20 Central sensitization following nerve injury: molecular mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:277-291. [PMID: 18808842 DOI: 10.1016/s0072-9752(06)80024-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
38
|
Cata JP, Weng HR, Chen JH, Dougherty PM. Altered discharges of spinal wide dynamic range neurons and down-regulation of glutamate transporter expression in rats with paclitaxel-induced hyperalgesia. Neuroscience 2006; 138:329-38. [PMID: 16361064 DOI: 10.1016/j.neuroscience.2005.11.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2004] [Revised: 10/17/2005] [Accepted: 11/08/2005] [Indexed: 01/06/2023]
Abstract
Changes in the signaling of wide dynamic range neurons and the expression of glutamate transporters in the lumbar spinal dorsal horn of rats with Taxol-induced hyperalgesia are detailed in this report. Deep spinal lamina neurons have significantly increased spontaneous activity and after-discharges to noxious mechanical stimuli, increased responses to both skin heating and cooling, and increased after-discharges and abnormal windup to transcutaneous electrical stimuli. The expression of glutamate transporter proteins in the dorsal horn is decreased at the time point corresponding to the physiological changes. These results suggest a state of increased excitability develops in spinal pain-signaling neurons as a consequence of decreased glutamate clearance. These changes in dorsal horn neurobiology likely in turn contribute to the hyper-responsiveness to sensory stimuli seen in animals treated with Taxol and may play a role in the pain seen in cancer patients receiving Taxol.
Collapse
Affiliation(s)
- J P Cata
- Department of Anesthesiology and Pain Research, Division of Anesthesiology and Critical Care Medicine, University of Texas-M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
39
|
Ossipov MH, Porreca F. Chapter 14 Descending excitatory systems. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:193-210. [PMID: 18808836 DOI: 10.1016/s0072-9752(06)80018-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Zou X, Lin Q, Willis WD. Effect of protein kinase C blockade on phosphorylation of NR1 in dorsal horn and spinothalamic tract cells caused by intradermal capsaicin injection in rats. Brain Res 2004; 1020:95-105. [PMID: 15312791 DOI: 10.1016/j.brainres.2004.06.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2004] [Indexed: 10/26/2022]
Abstract
We have previously reported that protein kinase A (PKA) is involved in the phosphorylation of NR1 subunits of N-methyl-d-aspartate (NMDA) receptors in dorsal horn neurons after intradermal injection of capsaicin (CAP). To see if protein kinase C (PKC) also participates in the phosphorylation of NR1, we used electron microscopic techniques to determine further where the phosphorylated NR1 subunits (pNR1) are expressed in the spinothalamic tract (STT) cells and immunohistochemistry to examine whether a PKC inhibitor, chelerythrine chloride, blocks the enhanced phosphorylation of NR1 on serine 896. The pNR1 subunits were in the soma and dendrites of STT cells and in presynaptic endings. Western blots showed that pretreatment with the PKC inhibitor caused a decrease in CAP-induced phosphorylation of NR1 protein. In immunofluorescence staining, the number of pNR1-like immunoreactive neurons was significantly decreased on the side ipsilateral to the injection when chelerythrine chloride was administered intrathecally before CAP injection. In addition, when STT cells were labeled by microinjection of the retrograde tracer, fluorogold (FG), into the thalamus, we found that the proportion of p-NR1-LI STT cells was markedly reduced after PKC inhibition. Combined with our previous findings, these results strongly suggest that NR1 subunits in spinal dorsal horn neurons are phosphorylated following CAP injection, and this phosphorylation is catalyzed by PKC, as well as by PKA.
Collapse
Affiliation(s)
- Xiaoju Zou
- Department of Anatomy and Neurosciences, Marine Biomedical Institute, The University of Texas Medical Branch, Galveston, TX 77555-1069, USA
| | | | | |
Collapse
|
41
|
Abstract
The generation of neuropathic pain is a complex phenomenon involving a process of peripheral and central sensitization producing enhanced transmission of nociceptive inputs to the brain associated with the loss of discriminatory processing of noxious and innocuous stimuli. This increased flow of abnormally processed nociceptive inputs to the brain may overcome the ability of descending modulatory pathways to produce analgesia, causing further worsening of the pain. Several crucial locations involved in the physiologic generation of pain inputs (eg, peripheral nociceptors, dorsal horns, thalamus, cortex) show evidence of functional reorganization and altered nociceptive processing in association with chronic pain. These locations present the best targets for therapeutic intervention, including systemic administration of drugs able to counteract the chemical storm induced by neural injuries in the nociceptive afferents and dorsal horns, or for more focused intervention, such as neuroablative procedures; intrathecal drug delivery; and spinal cord, deep brain, or motor cortex stimulation.
Collapse
Affiliation(s)
- Pantaleo Romanelli
- Epilepsy Surgery Unit, Department of Neurosurgery, Neuromed IRCCS, Pozzilli, Italy.
| | | |
Collapse
|
42
|
Argoff CE, Katz N, Backonja M. Treatment of postherpetic neuralgia: a review of therapeutic options. J Pain Symptom Manage 2004; 28:396-411. [PMID: 15471658 DOI: 10.1016/j.jpainsymman.2004.01.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2004] [Indexed: 11/22/2022]
Abstract
Postherpetic neuralgia (PHN) is a disabling consequence of the reactivation of the varicella zoster infection. The observation that patients with PHN experience various types of pain suggests that multiple pathophysiologic mechanisms are involved, which may include the peripheral and central nervous systems. A reasonable initial strategy would involve selecting from among multiple agents that have complementary mechanisms of action and that have been proven effective in controlled clinical trials, such as the lidocaine patch 5%, gabapentin, tricyclic antidepressants, and opioids. Based on initial assessment and ongoing reassessment, sequential trials should be undertaken until adequate pain relief is achieved. This may ultimately lead to therapy with more than one medication. Safety and tolerability are important considerations in choosing initial therapy, particularly in older patients. Physicians can either add another agent to the current regimen or switch to a new type of monotherapy if there is inadequate response to initial therapy. Alternative therapies, (i.e., ketamine, intrathecal corticosteroid injections) have not been adequately studied. Well-designed, multicenter, controlled clinical trials are needed to develop a treatment algorithm that provides an evidence-based, rational approach to treating PHN.
Collapse
Affiliation(s)
- Charles E Argoff
- Cohn Pain Management Center, North Shore University Hospital, Cohn Pain Management Center, Bethpage, New York 11714, USA
| | | | | |
Collapse
|
43
|
Palecek J, Neugebauer V, Carlton SM, Iyengar S, Willis WD. The effect of a kainate GluR5 receptor antagonist on responses of spinothalamic tract neurons in a model of peripheral neuropathy in primates. Pain 2004; 111:151-61. [PMID: 15327819 DOI: 10.1016/j.pain.2004.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2003] [Revised: 05/21/2004] [Accepted: 06/07/2004] [Indexed: 10/26/2022]
Abstract
The responses of antidromically identified spinothalamic tract (STT) neurons to mechanical and thermal stimuli were compared in anesthetized normal and neuropathic monkeys before and after administration of a GluR5 kainate receptor antagonist (LY382884) into the spinal cord dorsal horn through a microdialysis fiber. Peripheral neuropathy was induced by tight ligation of the L7 spinal nerve 13-15 days prior to the experiment. STT neurons recorded in the animals with neuropathy showed increased responsiveness to weak mechanical stimuli and to heating and cooling of the skin compared to STT cells in normal animals. In both normal and the neuropathic monkeys the responses of the STT neurons to mechanical and thermal stimuli were attenuated by LY382884 application in a concentration-dependent manner. Intraspinal application of LY382884 in the neuropathic animals led to a potent reduction of those responses of the STT neurons that were aggravated by the peripheral neuropathy (weak mechanical, heat and innocuous cooling stimuli). These results suggest that kainate receptors are involved in synaptic activation of STT cells in the normal state and may also play an important role in pathological pain states such as peripheral neuropathy in primates. Kainate receptor antagonists could thus be useful for the treatment of certain forms of allodynia and hyperalgesia.
Collapse
Affiliation(s)
- J Palecek
- Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
44
|
Lancelotta MP, Sheth RN, Meyer RA, Belzberg AJ, Griffin JW, Campbell JN. Severity and duration of hyperalgesia in rat varies with type of nerve lesion. Neurosurgery 2004; 53:1200-8; discussion 1208-9. [PMID: 14580288 DOI: 10.1227/01.neu.0000089482.80879.9a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2003] [Accepted: 05/21/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To learn how lesions with differing capacity for nerve regeneration affect the severity and duration of hyperalgesia in an animal model of neuropathic pain. METHODS Three groups of rats were studied: 1). L5 nerve root crush (favorable for regeneration); 2). L5 root ligation and section; and 3). sham-operated group. An experimenter who did not know the rats' groups tested the animals for hyperalgesia to mechanical and cold stimuli. RESULTS Measures of adverseness of mechanical and cooling stimuli for the crush group and ligation/cut groups were significantly higher than for the sham-operated group (P < 0.001 for both) for the first 30 days after lesioning. By 40 days, the crush group recovered from mechanical hyperalgesia, whereas the ligation/cut group continued to have significant hyperalgesia. At this time, both lesion groups displayed hyperalgesia to the cooling stimulus (P < 0.001), but the hyperalgesia in the ligation/cut group was significantly greater (P < 0.01). No recovery from cooling hyperalgesia was evident during the 54-day period of observation. Histological studies of the sciatic nerve indicated higher numbers of regenerating fibers in the crush group compared with the ligation/cut group. CONCLUSION This study demonstrates that axotomy, regardless of how it is induced, produces hyperalgesia to both mechanical and cold stimuli. However, the lesion that favors regeneration is associated with earlier signs of recovery from mechanical hyperalgesia and less severe signs of cooling hyperalgesia. The data support the hypothesis that inputs from the injured afferents play an ongoing role in neuropathic pain from nerve injury. Nerve ligation induces more severe and more sustained behavioral signs of pain than nerve crush.
Collapse
Affiliation(s)
- Mary Pat Lancelotta
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-7509, USA
| | | | | | | | | | | |
Collapse
|
45
|
Patte-Mensah C, Kappes V, Freund-Mercier MJ, Tsutsui K, Mensah-Nyagan AG. Cellular distribution and bioactivity of the key steroidogenic enzyme, cytochrome P450side chain cleavage, in sensory neural pathways. J Neurochem 2003; 86:1233-46. [PMID: 12911631 DOI: 10.1046/j.1471-4159.2003.01935.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neurosteroids are steroids produced within the nervous system. Based on behavioural responses evoked in animals by synthetic steroid injections, several studies suggested neurosteroid involvement in important neurophysiological processes. These observations should be correlated only to neuroactive effects of the injected steroids. Neurosteroids mostly control the CNS activity through allosteric modulation of neurotransmitter receptors within concentration ranges used by neurotransmitters themselves. Therefore, neurosteroid production within pathways controlling a neurophysiological process is necessary to consider neurosteroid involvement in that process. Because of the increasing speculation about pain modulation by neurosteroids based on pharmacological observations, we decided to clarify the situation by investigating neurosteroidogenesis occurrence in sensory pathways, particularly in nociceptive structures. We studied the presence and activity of cytochrome P450side chain cleavage (P450scc) in rat pain pathways. P450scc-immunoreactive cells were localized in dorsal root ganglia (DRG), spinal cord (SC) dorsal horn, nociceptive supraspinal nuclei (SSN) and somatosensory cortex. Incubation of DRG, SSN or SC tissue homogenates with [3H]cholesterol yielded the formation of radioactive metabolites including [3H]pregnenolone of which the synthesis was reduced in presence of aminogluthetimide, a P450scc inhibitor. These first neuroanatomical and neurochemical results demonstrate the occurrence of neurosteroidogenesis in nociceptive pathways and strongly suggest that neurosteroids may control pain mechanisms.
Collapse
Affiliation(s)
- Christine Patte-Mensah
- Laboratoire de Neurophysiologie Cellulaire et Intégrée, Unité Mixte de Recherche 7519-Centre National de la Recherche Scientifique, Université Louis Pasteur, 67084 Strasbourg Cedex, France
| | | | | | | | | |
Collapse
|
46
|
Hains BC, Johnson KM, Eaton MJ, Willis WD, Hulsebosch CE. Serotonergic neural precursor cell grafts attenuate bilateral hyperexcitability of dorsal horn neurons after spinal hemisection in rat. Neuroscience 2003; 116:1097-110. [PMID: 12617951 DOI: 10.1016/s0306-4522(02)00729-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemisection of the rat spinal cord at thoracic level 13 provides a model of spinal cord injury that is characterized by chronic pain attributable to hyperexcitability of dorsal horn neurons. Presuming that this hyperexcitability can be explained in part by interruption of descending inhibitory modulation by serotonin, we hypothesized that intrathecal transplantation of RN46A-B14 serotonergic precursor cells, which secrete serotonin and brain-derived neurotrophic factor, would reduce this hyperexcitability by normalizing the responses of low-threshold mechanoreceptive, nociceptive-specific, and multireceptive dorsal horn neurons. Three groups (n=45 total) of 30-day-old male Sprague-Dawley rats underwent thoracic level 13 spinal hemisection, after which four weeks were allowed for development of allodynia and hyperalgesia. The three groups of animals received transplants of no cells, 10(6) RN46A-V1 (vector-only) or 10(6) RN46A-B14 cells at lumbar segments 2-3. Electrophysiological experiments were done two weeks later. Low-threshold mechanoreceptive, nociceptive-specific, and multireceptive cells (n=394 total) were isolated at depths of 1-300 and 301-1000 micro in the lumbar enlargement. Responses to innocuous and noxious peripheral stimuli were characterized, and analyses of population responses were performed. Compared with normal animals, dorsal horn neurons of all types in hemisected animals showed increased responsiveness to peripheral stimuli. This was true for neurons on both sides of the spinal cord. After hemisection, the proportion of neurons classified as multireceptive cells increased, and interspike intervals of spontaneous discharges became less uniform after hemisection. Transplantation of RN46A-B14 cells restored evoked responses to near-control levels, normalized background activity, and returned the proportion of multireceptive cells to the control level. Restoration of normal activity was reversed with methysergide.These electrophysiological results corroborate anatomical and behavioral studies showing the effectiveness of serotonergic neural precursors in correcting phenomena associated with chronic central pain following spinal cord injury, and provide mechanistic insights regarding mode of action.
Collapse
Affiliation(s)
- B C Hains
- Department of Anatomy and Neurosciences, and Marine Biomedical Institute, University of Texas Medical Branch, Galveston, TX 77555-1043, USA
| | | | | | | | | |
Collapse
|
47
|
Hains BC, Willis WD, Hulsebosch CE. Serotonin receptors 5-HT1A and 5-HT3 reduce hyperexcitability of dorsal horn neurons after chronic spinal cord hemisection injury in rat. Exp Brain Res 2003; 149:174-86. [PMID: 12610685 DOI: 10.1007/s00221-002-1352-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2002] [Accepted: 11/21/2002] [Indexed: 01/21/2023]
Abstract
Spinal cord injury (SCI) results in abnormal pain syndromes in humans. In a rodent model of SCI, T13 spinal hemisection results in allodynia and hyperalgesia due in part to interruption of descending pathways, including serotonergic (5-HT) systems, that leads to hyperexcitability of dorsal horn neurons. To characterize further the role of 5-HT and 5-HT receptor subtypes 5-HT(1A) and 5-HT(3) in neuronal activation after hemisection, we have examined the responsiveness of dorsal horn neurons to a variety of innocuous and noxious peripheral stimuli. Male Sprague-Dawley rats, 150-175 g, were spinally hemisected (n=40) at T13 and allowed 4 weeks for development of mechanical allodynia and thermal hyperalgesia. Animals then underwent electrophysiologic recording and the results were compared with those from sham controls (n=15). Evoked responses of convergent dorsal horn neurons (n=224 total) at L3-L5 to innocuous and noxious peripheral stimuli were characterized after administration of vehicle, 5-HT (25, 50, 100, and 200 microg), 5-HT (100 microg) in conjunction with the selective 5-HT(1A) antagonist WAY 100135 (100 microg), the 5-HT(3) antagonist MDL 72222 (100 microg), the selective 5-HT(1A) agonist 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT, 150 microg), or the 5-HT(3) agonist 2-Me-5HT (75 microg), with or without pretreatment with antagonists; all treatments were delivered topically onto the cord adjacent to the recording electrode. In hemisected animals, increased responsiveness of convergent cells to all peripheral stimuli was observed bilaterally when compared to controls. No changes in ongoing background activity were present. In control animals, only the highest dose of 5-HT (200 microg) was sufficient to reduce evoked activity, whereas in hemisected animals a concentration-dependent decrease in response was observed. In hemisected animals, both 5-HT(1A) and 5-HT(3) receptor antagonism reduced the effectiveness of 5-HT, restoring elevated evoked activity by up to 70% at the doses tested. Administration of 5-HT(1A) and 5-HT(3) receptor agonists also decreased hyperexcitability, effects prevented by pretreatment with corresponding antagonists. These results demonstrate the development of denervation supersensitivity to 5-HT following SCI, corroborate behavioral studies showing the effectiveness of 5-HT in reducing allodynia and hyperalgesia after SCI, and contribute to a mechanistic understanding of the role of 5-HT receptor subtypes in chronic central pain.
Collapse
Affiliation(s)
- Bryan C Hains
- Department of Anatomy and Neurosciences, and Marine Biomedical Institute, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-043, USA
| | | | | |
Collapse
|
48
|
Wall JT, Xu J, Wang X. Human brain plasticity: an emerging view of the multiple substrates and mechanisms that cause cortical changes and related sensory dysfunctions after injuries of sensory inputs from the body. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2002; 39:181-215. [PMID: 12423766 DOI: 10.1016/s0165-0173(02)00192-3] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Injuries of peripheral inputs from the body cause sensory dysfunctions that are thought to be attributable to functional changes in cerebral cortical maps of the body. Prevalent theories propose that these cortical changes are explained by mechanisms that preeminently operate within cortex. This paper reviews findings from humans and other primates that point to a very different explanation, i.e. that injury triggers an immediately initiated, and subsequently continuing, progression of mechanisms that alter substrates at multiple subcortical as well as cortical locations. As part of this progression, peripheral injuries cause surprisingly rapid neurochemical/molecular, functional, and structural changes in peripheral, spinal, and brainstem substrates. Moreover, recent comparisons of extents of subcortical and cortical map changes indicate that initial subcortical changes can be more extensive than cortical changes, and that over time cortical and subcortical extents of change reach new balances. Mechanisms for these changes are ubiquitous in subcortical and cortical substrates and include neurochemical/molecular changes that cause functional alterations of normal excitation and inhibition, atrophy and degeneration of normal substrates, and sprouting of new connections. The result is that injuries that begin in the body become rapidly further embodied in reorganizational make-overs of the entire core of the somatosensory brain, from peripheral sensory neurons to cortex. We suggest that sensory dysfunctions after nerve, root, dorsal column (spinal), and amputation injuries can be viewed as diseases of reorganization in this core.
Collapse
Affiliation(s)
- J T Wall
- Cellular and Molecular Neurobiology Program, Medical College of Ohio, Toledo 43614-5804, USA.
| | | | | |
Collapse
|
49
|
Suzuki R, Dickenson AH. Nerve injury-induced changes in opioid modulation of wide dynamic range dorsal column nuclei neurones. Neuroscience 2002; 111:215-28. [PMID: 11955724 DOI: 10.1016/s0306-4522(01)00617-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the present study we investigated the effects of spinal morphine on the electrically and naturally evoked responses of gracile nuclei neurones in a rat model of neuropathy, induced by the tight ligation of lumbar L5/6 spinal nerves. Two weeks after surgery, animals were prepared for electrophysiological recordings and neuronal responses were characterised to a range of controlled natural (brush, low- and high-intensity von Frey filaments, heat 45 degrees C) and peripheral electrical stimuli. Morphine (0.1, 0.25, 1 and 5 microg) was applied spinally and its effect was compared to that in sham-operated or naive animals. Following surgery, all neuropathic rats exhibited signs of mechanical allodynia. Nerve injury induced a significant increase in the receptive field size of gracile nuclei neurones, and also produced a non-significant increase in the proportion and level of spontaneous activity in these neurones. The baseline electrical and natural evoked responses remained unaltered. Spinal morphine reduced both the Adelta-fibre- and C-fibre-evoked responses of gracile nuclei neurones, and similarly inhibited the heat-evoked responses of neuropathic, sham-operated and naive rats. Morphine, however, produced only minor reductions (<30% inhibition of pre-drug control responses) of the Abeta-fibre- and brush-evoked responses of gracile nuclei neurones. These drug effects were similar in all animal groups. In complete contrast, morphine produced a marked inhibition of the low-intensity punctate mechanical evoked responses (von Freys 2 and 9 g) after nerve injury, an effect that was totally lacking in the sham-operated or naive animal groups. This dramatic shift was selective for the low-intensity punctate mechanical stimuli and such an effect was not seen with the noxious mechanical punctate stimulus (von Frey 75 g) where there was a modest inhibition in all groups. Our results suggest that there is plasticity in the opioid modulation of dorsal column projection pathways following spinal nerve ligation and these alterations appear to interact with sensory pathways conveying low-threshold punctate stimuli.
Collapse
Affiliation(s)
- R Suzuki
- Department of Pharmacology, University College London, Gower Street, WC1E 6BT, London, UK.
| | | |
Collapse
|
50
|
Andrew D, Craig AD. Responses of spinothalamic lamina I neurons to maintained noxious mechanical stimulation in the cat. J Neurophysiol 2002; 87:1889-901. [PMID: 11929909 DOI: 10.1152/jn.00577.2001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Noxious mechanical stimuli that are maintained for minutes produce a continuous sensation of pain in humans that augments during the stimulus. It has recently been shown with systematic force-controlled stimuli that, while all mechanically responsive nociceptors adapt to these stimuli, the basis for such pain can be ascribed to A-fiber rather than C-fiber nociceptors, based on distinctions in their respective response profiles and stimulus-response functions. The present experiments investigated whether similar distinctions could be made in subsets of nociceptive lamina I spinothalamic tract (STT) neurons using similar maintained stimuli. Twenty-eight lamina I STT neurons in the lumbosacral dorsal horn of barbiturate-anesthetized cats were tested with noxious mechanical stimuli applied with a probe of 0.1 mm(2) contact area at forces of 25, 50, and 100 g for 2 min. The neurons were classified as nociceptive-specific (NS, n = 14) or polymodal nociceptive (HPC, n = 14) based on their responses to quantitative thermal stimuli. The NS neurons had greater responses and showed less adaptation than the HPC neurons in response to these stimuli, and they encoded stimulus intensity better. Comparison of the normalized response profiles of all 28 nociceptive lamina I STT neurons, independent of cell classification, revealed 2 subgroups that differed significantly: "Maintained" cells with responses that remained above 50% of the initial peak rate during stimulation and "Adapting" cells with responses that quickly declined to <50%. The Maintained neurons encoded the intensity of the mechanical stimuli better than the Adapting neurons, based on ratiometric functions. A k-means cluster analysis of all 28 cells distinguished the identical two subgroups. These categories corresponded closely to the NS and HPC categories: Maintained cells were mostly NS neurons (10 NS, 3 HPC), and Adapting cells were mostly HPC neurons (4 NS, 11 HPC). Thus the present data are consistent with the distinctions between A-fiber and C-fiber nociceptors observed previously, because A-fiber nociceptors are the predominant input to NS lamina I STT neurons and C-fiber nociceptors are the predominant input to HPC neurons. These findings support the view that NS, but perhaps not HPC, lamina I STT neurons have a role in the pain caused by maintained mechanical stimuli and contribute to the sensations of "first" pain and "sharpness." Nonetheless, none of the units studied showed increasing responses during the stimuli, suggesting a role for other ascending neurons or forebrain integration in the augmenting pain produced by maintained mechanical stimulation.
Collapse
Affiliation(s)
- D Andrew
- Atkinson Pain Research Laboratory, Division of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA
| | | |
Collapse
|