Dissecting out mechanisms responsible for peripheral neuropathic pain: Implications for diagnosis and therapy

Similar documents
Seizure: the clinical manifestation of an abnormal and excessive excitation and synchronization of a population of cortical

A role for uninjured afferents in neuropathic pain

Pathophysiological Classification of Pain

Special Issue on Pain and Itch

Introduction to some interesting research questions: Molecular biology of the primary afferent nociceptor

Peripheral nerve injury alters excitatory synaptic transmission in lamina II of the rat dorsal horn

Pain Mechanisms. Prof Michael G Irwin MD, FRCA, FANZCA FHKAM Head Department of Anaesthesiology University of Hong Kong. The Somatosensory System

Pharmacology of Pain Transmission and Modulation

Update on the Neurophysiology of Pain Transmission and Modulation: Focus on the NMDA-Receptor

Brian Kahan, D.O. FAAPMR, DABPM, DAOCRM, FIPP Center for Pain Medicine and Physiatric Rehabilitation 2002 Medical Parkway Suite 150 Annapolis, MD

Spinal Cord Injury Pain. Michael Massey, DO CentraCare Health St Cloud, MN 11/07/2018

Pain Pathways. Dr Sameer Gupta Consultant in Anaesthesia and Pain Management, NGH

PAIN MANAGEMENT in the CANINE PATIENT

Sensory coding and somatosensory system

The Egyptian Journal of Hospital Medicine (January 2018) Vol. 70 (12), Page

Pain teaching. Muhammad Laklouk

Biomechanics of Pain: Dynamics of the Neuromatrix

Pathophysiology of Pain. Ramon Go MD Assistant Professor Anesthesiology and Pain medicine NYP-CUMC

Somatosensory Physiology (Pain And Temperature) Richard M. Costanzo, Ph.D.

Applied physiology: neuropathic pain

San Francisco Chronicle, June 2001

Venipuncture-induced neuropathic pain: the clinical syndrome, with comparisons to experimental nerve injury models

A Review of Neuropathic Pain: From Diagnostic Tests to Mechanisms

Neuropathic pain: aetiology, s y m p t o m s, m e c h a n i s m s, a n d m a n a g e m e n t

Neuropathic pain, pain matrix dysfunction, and pain syndromes

Enhanced formalin nociceptive responses following L5 nerve ligation in the rat reveals neuropathy-induced inflammatory hyperalgesia

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance

Cancer-induced bone pain

What it Takes to be a Pain

Somatic Sensation (MCB160 Lecture by Mu-ming Poo, Friday March 9, 2007)

SYLLABUS SPRING 2011 COURSE: NSC NEUROBIOLOGY OF PAIN

EDUCATION M.D., Peking Union Medical College, Beijing, China, 1999 B.S., Beijing University, College of Life Science, Beijing, China, 1994

Mechanical sensitization of cutaneous sensory fibers in the spared nerve injury mouse model

Receptors and Neurotransmitters: It Sounds Greek to Me. Agenda. What We Know About Pain 9/7/2012

Pathophysiology of Pain

From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain

ANAT2010. Concepts of Neuroanatomy (II) S2 2018

PAIN IS A SUBJECTIVE EXPERIENCE: It is not a stimulus. MAJOR FEATURES OF THE PAIN EXPERIENCE: Sensory discriminative Affective (emotional) Cognitive

Preemptive Analgesia: Does it Prevent Chronic Pain?

Pathophysiological Mechanisms of Neuropathic Pain

Chronic pain: We should not underestimate the contribution of neural plasticity. *Gwyn N Lewis 1, David A Rice 1,2

Mechanism of Pain Production

What is Pain? An unpleasant sensory and emotional experience associated with actual or potential tissue damage. Pain is always subjective

The anatomy and physiology of pain

Function of the Nervous System

The perception of pain is a complex process,

Functions of Nervous System Neuron Structure

Neuropathic Pain in Palliative Care

Proceedings of the World Small Animal Veterinary Association Sydney, Australia 2007

THE EFFECT OF VOLUNATARY EXERCISE ON NEUROPATHIC PAIN. Kevin L. Farmer

211MDS Pain theories

S E C T I O N I M E C H A N I S M S A N D E P I D E M I O L O G Y

STRUCTURAL ELEMENTS OF THE NERVOUS SYSTEM

Module H NERVOUS SYSTEM

Pre-op Interventions to Mitigate Post-op Acute and Chronic Pain

Psychophysical laws. Legge di Fechner: I=K*log(S/S 0 )

Modulation of TRP channels by resolvins in mouse and human

ANATOMY AND PHYSIOLOGY OF NEURONS. AP Biology Chapter 48

Synaptic plasticityhippocampus. Neur 8790 Topics in Neuroscience: Neuroplasticity. Outline. Synaptic plasticity hypothesis

EE 791 Lecture 2 Jan 19, 2015

Received 26 April 2004; revised 21 August 2004; accepted 20 September 2004 Available online 19 November 2004

Neurons, Synapses, and Signaling

Introduction to Neurobiology

The biochemical origin of pain: The origin of all pain is inflammation and the inflammatory response: Inflammatory profile of pain syndromes

What Does the Mechanism of Spinal Cord Stimulation Tell Us about Complex Regional Pain Syndrome?pme_

Chapter 7 Nerve Cells and Electrical Signaling

Name Biology 125 Midterm #2 ( ) Total Pages: 9

Physiology of synapses and receptors

10.1: Introduction. Cell types in neural tissue: Neurons Neuroglial cells (also known as neuroglia, glia, and glial cells) Dendrites.

Inherited erythromelalgia mutations in Na v 1.7

Nervous System. Master controlling and communicating system of the body. Secrete chemicals called neurotransmitters

Index. Dent Clin N Am 51 (2007) Note: Page numbers of article titles are in boldface type.

CHAPTER 4 PAIN AND ITS MANAGEMENT

Chapter 11: Functional Organization of Nervous Tissue

Potential for delta opioid receptor agonists as analgesics in chronic pain therapy

Corporate Medical Policy

Chapter 16. Sense of Pain

Outline. Animals: Nervous system. Neuron and connection of neurons. Key Concepts:

The Nervous System. Dr. ZHANG Xiong Dept. of Physiology ZJU School of Medicine.

Pain. Types of Pain. Types of Pain 8/21/2013

What is pain?: An unpleasant sensation. What is an unpleasant sensation?: Pain. - Aristotle.

Hole s Human Anatomy and Physiology Eleventh Edition. Chapter 10

The Nervous System. Nervous System Functions 1. gather sensory input 2. integration- process and interpret sensory input 3. cause motor output

Analgesic effects of the COX-2 inhibitor parecoxib on surgical pain through suppression of spinal ERK signaling

Bi/CNS/NB 150: Neuroscience. November 11, 2015 SOMATOSENSORY SYSTEM. Ralph Adolphs

ANAT2010. Concepts of Neuroanatomy (II) S2 2018

浙江大学医学院基础医学整合课程 各论 III. The Nervous System. Dr. ZHANG Xiong Dept. of Physiology ZJU School of Medicine

Neuroscience: Exploring the Brain, 3e. Chapter 4: The action potential

Shift 1, 8 July 2018, 09:30-13:00

Intracellular signalling cascades associated with TRP channels

Chapter 12 Nervous Tissue. Copyright 2009 John Wiley & Sons, Inc. 1

SOMATOSENSORY SYSTEMS AND PAIN

Department of Neurology/Division of Anatomical Sciences

Ligand-Gated Ion Channels

Somatosensation. Recording somatosensory responses. Receptive field response to pressure

References. Agarwal, N., Pacher, P., Tegeder, I., Amaya, F., Constantin, C. E., Brenner, G. J., et al. (2007).

5-Nervous system II: Physiology of Neurons

Outline. Neuron Structure. Week 4 - Nervous System. The Nervous System: Neurons and Synapses

Transcription:

Life Sciences 74 (2004) 2605 2610 www.elsevier.com/locate/lifescie Dissecting out mechanisms responsible for peripheral neuropathic pain: Implications for diagnosis and therapy Clifford J. Woolf* Neural Plasticity Research Group, Department of Anesthesia & Critical Care, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA Abstract Peripheral neuropathic pain, that clinical pain syndrome associated with lesions to the peripheral nervous system, is characterized by positive and negative symptoms. Positive symptoms include spontaneous pain, paresthesia and dysthesia, as well as a pain evoked by normally innocuous stimuli (allodynia) and an exaggerated or prolonged pain to noxious stimuli (hyperalgesia/hyperpathia). The negative symptoms essentially reflect loss of sensation due to axon/neuron loss, the positive symptoms reflect abnormal excitability of the nervous system. Diverse disease conditions can result in neuropathic pain but the disease diagnosis by itself is not helpful in selecting the optimal pain therapy. Identification of the neurobiological mechanisms responsible for neuropathic pain is leading to a mechanism-based approach to this condition, which offers the possibility of greater diagnostic sensitivity and a more rational basis for therapy. We are beginning to move from an empirical symptom control approach to the treatment of pain to one targeting the specific mechanisms responsible. This review highlights some of the mechanisms underlying neuropathic pain and the novel targets they reveal for future putative analgesics. D 2004 Elsevier Inc. All rights reserved. Keywords: Peripheral neuropathic pain; Primary sensory neurons; Ectopic excitability; Phenotypic switch; Sensory neuron loss; Central sensitization; Disinhibition Mechanisms of peripheral neuropathic pain Multiple mechanisms contribute to the neuropathic pain syndrome. These include changes in the peripheral nervous system, spinal cord brainstem and brain. The temporal profiles of the mechanisms * Corresponding author. Neural Plasticity Research Group, Department of Anesthesia and Critical Care, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA. Tel.: +1-617-724-3622; fax: +1-617-724-3632. E-mail address: cwoolf@partners.org (C.J. Woolf). 0024-3205/$ - see front matter D 2004 Elsevier Inc. All rights reserved. doi:10.1016/j.lfs.2004.01.003

2606 C.J. Woolf / Life Sciences 74 (2004) 2605 2610 differ, some are transient, some require the presence of ongoing peripheral pathology for their maintenance, and others produce persistent or autonomous changes in the operation of the nervous system. In a given patient multiple mechanisms may co-exist, there may be a temporal evolution from one mechanism to another over the course of the natural history of the disease, or a single mechanism may dominate. The etiological factors responsible for driving the mechanisms are not disease specific (Woolf et al., 1998). Patients with postherpetic neuralgia, for example, may have multiple mechanisms and may share mechanisms with patients with painful peripheral diabetic neuropathy (Koltzenburg, 1998; Woolf and Mannion, 1999). We face three major challenges in studying neuropathic pain mechanisms. The first is, what are the neurobiological mechanisms responsible for the pain (Devor and Seltzer, 1999; Scholz and Woolf, 2002)? The second is, how can we identify which mechanism operates in patients to produce their pain (Galer and Jensen, 1997; Rowbotham and Fields, 1996; Woolf and Decosterd, 1999)? The third is to develop pharmacological tools that are targeted specifically at the mechanisms and enable their disruption (Sindrup and Jensen, 1999; Woolf and Max, 2001). The unifying feature of peripheral neuropathic pain is pain in the presence of a lesion, damage or disruption to some component of primary sensory neurons. The lesion may be in a peripheral nerve, the dorsal root ganglion or a dorsal root and may be the consequence of trauma, compression, tumor invasion, ischemia, inflammation, metabolic disturbances, nutritional deficits, cytotoxic agents and degenerative disorders (Woolf and Mannion, 1999). These factors act to interfere with or damage some part of a primary sensory neuron, its peripheral axon, cell body or central axon. A change in the function of the somatosensory system with no evidence of a lesion to the PNS, as in fibromyalgia, migraine, irritable bowel syndrome, should not strictly speaking, be considered neuropathic pain. Disruption of the continuity of a primary sensory neuron with its peripheral target or, a loss of the neuron due cell death, will result in a loss of sensory inflow and some detectable sensory impairment. Positive symptoms result from changes in the injured primary sensory neurons but also in neighboring non-injured sensory neurons, as well as transynaptic changes in neurons at multiple levels of the CNS. What are these changes and how do they produce pain? I will briefly survey a few of the key mechanisms to illustrate how identifying these represent a major step forward in our understanding of neuropathic pain. Ectopic excitability Injured and neighboring non-injured sensory neurons can develop a change in their excitability sufficient to generate pacemaker-like potentials, which evoke ectopic action potential discharges, a sensory inflow independent of any peripheral stimulus (Devor and Seltzer, 1999; Liu et al., 2002). These changes may manifest at the site of the injury, at the neuroma, and in the DRG. Ectopic input is most prominent in A fibers but also occurs to a more limited extent in cells with unmyelinated axons (Devor and Seltzer, 1999). Three factors appear to be responsible, upregulation of voltage gated sodium channels, including Nav1.3 and Nav1.8, down regulation of potassium channels, and possibly a reduction in threshold of Trp transducer heat sensitive channels so that they can be activated at body temperature (Waxman et al., 1999). The ectopic activity may directly initiate spontaneous sensations; paresthesia, dysthethesia and burning pain. In addition the spontaneous inflow may generate activitydependent changes in excitability of central neurons (central sensitization). Potential treatment options aimed at ectopic activity include sodium channel blockers (ideally use-dependent blockers targeted at the

C.J. Woolf / Life Sciences 74 (2004) 2605 2610 2607 specific channels involved), potassium channel openers, Trp channel blockers, and treatment targeted at the mechanisms responsible for the alterations in the density, distribution or kinetics of these channels, blocking the alterations in the transcription, post-translational changes and trafficking of the ion channels after nerve injury. Phenotypic switch Differentiated neurons are characterized by expression of a large number of specific genes enabling the cells to carry out their particular functions. For primary sensory neurons these include those genes that enable transduction, conduction, and synaptic transmission as well as many housekeeping and cytoskeletal genes. After peripheral nerve injury there is a surprisingly large change in the levels of transcripts, several hundred genes are either up or down regulated (Costigan et al., 2002; Xiao et al., 2002). These include regeneration-associated genes, survival factors and many genes that determine the function or malfunction of the neurons. A consequence of these changes are alterations in the excitability of the neurons (ectopic excitability, see above), as well as their transduction, and transmitter properties. Some changes include a switch in the phenotype of the neurons. For example, the neuromodulators BDNF and substance P are normally expressed only in C-fibers, but begin after peripheral nerve injury to be expressed in A fiber neurons (Noguchi et al., 1995; Fukuoka et al., 2001). This may mean that these fibers acquire the capacity to produce central changes such as central sensitization, which is normally produced only by C-fibers (Decosterd et al., 2002). The identification of the altered expression profile of sensory neurons after nerve injury is revealing many exciting new putative targets for novel analgesics and helps explain the differential action of some existing analgesics. The a2y calcium channel subunit, for example, is markedly upregulated after nerve injury (Luo et al., 2001; Costigan et al., 2002), and this may contribute to the analgesic action of gabapentinoids in neuropathic pain. Primary sensory degeneration Peripheral nerve injury disrupts the contact of the cell bodies of DRG neurons with their peripheral targets. These targets are a source of growth factors such as NGF or GDNF. Following peripheral axonal injury there are over weeks, atrophic changes in the injured neurons, a reduction in axon caliber, a decrease in the size of the cell body and a loss of the contact that the central terminals of the afferents make with spinal cord neurons. Later, some months after the nerve injury some neurons begin to die, most of which are C-fibers (Tandrup et al., 2000). Although most treatment of neuropathic pain is aimed at reducing the positive symptoms, loss of neurons and the resulting imbalance of sensory inflow may contribute to the abnormal sensations. We need to evaluate what is responsible for nerve-injury induced sensory neuron loss and if preventing this is beneficial. Central sensitization Central sensitization represents a state of heightened sensitivity of dorsal horn neurons such that their threshold of activation is reduced, and their responsiveness to synaptic inputs is augmented, essentially

2608 C.J. Woolf / Life Sciences 74 (2004) 2605 2610 the gain of the system is increased (Woolf and Salter, 2000). There are two forms of central sensitization; an activity-dependent form that is rapidly induced with in seconds by afferent activity in nociceptors and which produces changes in synaptic efficacy that last for tens of minutes as a result of the phosphorylation and altered trafficking of voltage- and ligand-gated ion channel receptors, and a transcription-dependent form that takes some hours to be induced but outlast the initiating stimulus for prolonged periods. Under normal conditions the activity-dependent form of central sensitization is produced only following the activation of small caliber Ay and C fiber afferents by a noxious or tissue damaging stimulus (Woolf, 1983; Woolf and Wall, 1986). After peripheral nerve injury C-fiber input may arise spontaneously and drive central sensitization. In addition, the phenotypic changes that occur in A fibers after nerve injury (see above) enable them now to drive central sensitization and repeated light touch can after nerve injury begin to produce central sensitization (Decosterd et al., 2002). The activitydependent form of central sensitization is responsible for generating secondary pinprick hyperalgesia and dynamic tactile allodynia (Campbell et al., 1988; Koltzenburg et al., 1992). It obviously represents a major target for drug intervention, and the NMDA and AMPA receptors have been shown in may preclinical and clinical studies to have a major role, including in patients with neuropathic pain (Felsby et al., 1996). The problem is that these receptors are so widespread that there is insufficient therapeutic index to produce analgesia without significant CNS side effects. There may be other ways direct and indirect to reduce activity-dependent central sensitization. Indirect approaches could include reducing ectopic activity and preventing phenotypic switches. More direct approaches could include reducing transmitter release with calcium channel blockers (N-type) and a2y binding drugs, reducing posttranslational changes with PKA, PKC, src or MAPK inhibitors, potassium channel openers, as well as antagonists for ligand-gated and G protein coupled receptors. Transcription-dependent central sensitization has been studied in the context of peripheral inflammation where changes in BDNF, TrB, substance P, NK1, dynorphin and Cox2 are well described (Neumann et al., 1996; Mannion et al., 1999; Ji et al., 2002). Much less is know about when, how and for how long this form of plasticity manifests in the dorsal horn after peripheral nerve injury, and the extent to which it contributes to neuropathic pain, this is an exciting challenge. There is certainly evidence that microglial activation may be involved (Jin et al., 2003; Watkins et al., 2001). Disinhibition The balance of excitatory and inhibitory influences on neurons plays a major role in determining information flow through CNS circuits. Increases in excitation produced by increased inputs (ectopic activity) and increased responsiveness (central sensitization) shift the balance to increased excitability, which can manifest as spontaneous or evoked pain. A reduction in inhibition can have a very similar net result. Pharmacologically, blocking GABA or glycine-mediated inhibition produces a pattern of pain hypersensitivity very similar to that of neuropathic pain with very prominent tactile allodynia (Sivilotti and Woolf, 1994), and GABA blockade recruits previously absent Ah fiber inputs to lamina II cells, effectively uncovering a previously silent synaptic pathway (Baba et al., 2003). It turns out that partial nerve injury also reduces inhibition in the superficial dorsal horn with a selective loss of GABAergic inhibitory synaptic currents that is due to induction in GABAergic inhibitory interneurons of apoptosis (Moore et al., 2002). Peripheral nerve injury induces then, a transynaptic neural degeneration that results in a loss of function which contributes to abnormal pain sensitivity. The implications of this are

C.J. Woolf / Life Sciences 74 (2004) 2605 2610 2609 considerable. It may become possible to prevent after nerve injury, cell death in the dorsal horn and in this way abort some elements of neuropathic pain preventing its chronicity. This would represent a disease modifying approach to the condition rather than just symptom control. There remain many questions to be answered, what is responsible for the nerve injury induced cell death, what is the duration of the therapeutic window of opportunity, what is the best form of therapy to prevent the cell loss. Conclusion Dissecting out the mechanisms responsible for the production of neuropathic pain is helping both to explain how existing drugs produce their analgesic effects and is revealing novel biological and molecular targets which will lead to new drugs. Neuropathic pain constitutes a major clinical problem. Given the pace of recent advances in our understanding it is at last realistic to expect that new and improved therapy will become available to help control the syndrome. References Baba, H., Ji, R.R., Kohno, T., Moore, K.A., Ataka, T., Wakai, A., Okamoto, M., Woolf, C.J., 2003. Removal of GABAergic inhibition facilitates polysynaptic A fiber-mediated excitatory transmission to the superficial spinal dorsal horn. Mol. Cell. Neurosci. 24, 818 830. Campbell, J.N., Raja, S.N., Meyer, R.A., McKinnon, S.E., 1988. Myelinated afferents signal the hyperalgesia associated with nerve injury. Pain 32, 89 94. Costigan, M., Befort, K., Karchewski, L., Griffin, R.S., D Urso, D., Allchorne, A., Sitarski, J., Mannion, J.W., Pratt, R.E., Woolf, C.J., 2002. Replicate high-density rat genome oligonucleotide microarrays reveal hundreds of regulated genes in the dorsal root ganglion after peripheral nerve injury. BMC Neurosci. 3, 16. Decosterd, I., Allchorne, A., Woolf, C.J., 2002. Progressive tactile hypersensitivity after a peripheral nerve crush: non-noxious mechanical stimulus-induced neuropathic pain. Pain 100, 155 162. Devor, M., Seltzer, Z., 1999. Pathophysiology of damaged nerves in realtion to chronic pain. In: Wall, P.D., Melzack, R. (Eds.), Textbook of Pain, Churchill Livingstone, Edinburg, pp. 129 164. Felsby, S., Nielsen, J., Arendt-Nielsen, L., Jensen, T.S., 1996. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium cholride. Pain 64, 283 291. Fukuoka, T., Kondo, E., Dai, Y., Hashimoto, N., Noguchi, K., 2001. Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model. J. Neurosci. 21, 4891 4900. Galer, B.S., Jensen, M.P., 1997. Development and preliminary validation of a pain measure specific to neuropathic pain: the Neuropathic Pain Scale. Neurology 48, 332 338. Ji, R.R., Befort, K., Brenner, G.J., Woolf, C.J., 2002. ERK MAP Kinase Activation in Superficial Spinal Cord Neurons Induces Prodynorphin and NK-1 Upregulation and Contributes to Persistent Inflammatory Pain Hypersensitivity. J. Neurosci. 22, 478 485. Jin, S.X., Zhuang, Z.Y., Woolf, C.J., Ji, R.R., 2003. p38 Mitogen-Activated Protein Kinase Is Activated after a Spinal Nerve Ligation in Spinal Cord Microglia and Dorsal Root Ganglion Neurons and Contributes to the Generation of Neuropathic Pain. J. Neurosci. 23, 4017 4022. Koltzenburg, M., 1998. Painful neuropathies. Curr. Opin. Neurol. 11, 515 521. Koltzenburg, M., Wahren, L.K., Torebjork, H.E., 1992. Dynamic changes of mechanical hyperalgesia in neuropathic pain states and healthy subjects depend on the ongoing activity of unmyelinated nociceptive afferents. Pflugers. Arch. 420, R52. Liu, C.N., Devor, M., Waxman, S.G., Kocsis, J.D., 2002. Subthreshold oscillations induced by spinal nerve injury in dissociated muscle and cutaneous afferents of mouse DRG. J. Neurophysiol. 87, 2009 2017. Luo, Z.D., Chaplan, S.R., Higuera, E.S., Sorkin, L.S., Stauderman, K.A., Williams, M.E., Yaksh, T.L., 2001. Upregulation of

2610 C.J. Woolf / Life Sciences 74 (2004) 2605 2610 dorsal root ganglion (alpha)2(delta) calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J. Neurosci. 21, 1868 1875. Mannion, R.J., Costigan, M., Decosterd, I., Amaya, F., Ma, Q.P., Holstege, J.C., Ji, R.R., Acheson, A., Lindsay, R.M., Wilkinson, G.A., Woolf, C.J., 1999. Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc. Natl. Acad. Sci. U. S. A. 96, 9385 9390. Moore, K.A., Kohno, T., Karchewski, L.A., Scholz, J., Baba, H., Woolf, C.J., 2002. Partial peripheral nerve injury promotes a selective loss of GABAergic inhibition in the superficial dorsal horn of the spinal cord. J. Neurosci. 15, 6724 6731. Neumann, S., Doubell, T.P., Leslie, T.A., Woolf, C.J., 1996. Inflammatory pain hypersensitivity mediated by phenotypic switch in myelinated primary sensory neurones. Nature 384, 360 364. Noguchi, K., Kawai, Y., Fukuoka, T., Senba, E., Miki, K., 1995. Substance P induced by peripheral nerve injury in primary afferent sensory neurons and its effect on dorsal column nucleus neurons. J. Neurosci. 15, 7633 7643. Rowbotham, M.C., Fields, H.L., 1996. The relationship of pain, allodynia and thermal sensation in post-herpetic neuralgia. Brain 119 (Pt 2), 347 354. Scholz, J., Woolf, C.J., 2002. Can we conquer pain? Nat. Neurosci. 5, 1062 1067 (Suppl.). Sindrup, S.H., Jensen, T.S., 1999. Efficacy of pharmacological treatments of neuropathic pain: an update and effect related to mechanism of drug action. Pain 83, 389 400. Sivilotti, L.G., Woolf, C.J., 1994. The contribution of GABA A and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J. Neurophysiol. 72, 169 179. Tandrup, T., Woolf, C.J., Coggeshall, R.E., 2000. Delayed loss of small dorsal root ganglion cells after transection of the rat sciatic nerve. J. Comp. Neurol. 422, 172 180. Watkins, L.R., Milligan, E.D., Maier, S.F., 2001. Glial activation: a driving force for pathological pain. Trends Neurosci. 24, 450 455. Waxman, S.G., Dib-Hajj, S., Cummins, T.R., Black, J.A., 1999. Sodium channels and pain. Proc. Natl. Acad. Sci. U. S. A. 96, 7635 7639. Woolf, C.J., 1983. Evidence for a central component of post-injury pain hypersensitivity. Nature 306, 686 688. Woolf, C.J., Bennett, G.J., Doherty, M., Dubner, R., Kidd, B., Koltzenburg, M., Lipton, R., Loeser, J.D., Payne, R., Torebjork, E., 1998. Towards a mechanism-based classification of pain? Pain 77, 227 229. Woolf, C.J., Decosterd, I., 1999. Implications of recent advances in the understanding of pain pathophysiology for the assessment of pain in patients. Pain 82, 1 7. Woolf, C.J., Mannion, R.J., 1999. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet 353, 1959 1964. Woolf, C.J., Max, M.B., 2001. Mechanism-based pain diagnosis: issues for analgesic drug development. Anesthesiol. 95, 241 249. Woolf, C.J., Salter, M.W., 2000. Neuronal plasticity-increasing the gain in pain. Science 288, 1765 1768. Woolf, C.J., Wall, P.D., 1986. The relative effectiveness of C primary afferent fibres of different origins in evoking a prolonged facilitation of the flexor reflex in the rat. J. Neurosci. 6, 1433 1443. Xiao, H.S., Huang, Q.H., Zhang, F.X., Bao, L., Lu, Y.J., Guo, C., Yang, L., Huang, W.J., Fu, G., Xu, S.H., Cheng, X.P., Yan, Q., Zhu, Z.D., Zhang, X., Chen, Z., Han, Z.G., Zhang, X., 2002. Identification of gene expression profile of dorsal root ganglion in the rat peripheral axotomy model of neuropathic pain. Proc. Natl. Acad. Sci. U. S. A. 99, 8360 8365.