Journal of. The South African Chapter of the IASP ISSN Central sensitization: Implications for the diagnosis and treatment of pain

Size: px
Start display at page:

Download "Journal of. The South African Chapter of the IASP ISSN Central sensitization: Implications for the diagnosis and treatment of pain"

Transcription

1 Journal of ISSN The South African Chapter of the IASP Central sensitization: Implications for the diagnosis and treatment of pain Orofacial pain Pathophysiology of postoperative pain Volume 6 Number

2

3 Journal of The South African Chapter of the IASP Volume 6 Number 4 Editorial The IASP is always striving to improve knowledge concerning pain and pain therapy. One of the recent changes in their structure/curriculum was to decrease the time between World Congresses. Those of us who have been fortunate enough to attend these congresses can attest to the value of participation. Besides the normal abstract book or CD, attendees also used to receive a book titled Proceedings of the World Pain Congress. This is no longer the case. instead asked plenary speakers to prepare peer-reviewed articles review articles in which they summarise the current thoughts and evidence presented in one of their plenary lectures. These reviews will from now be published as a Special Issue of Pain. I have selected several articles that cover a range of practical topics that will be valuable to all of our members. Clifford Woolf needs no introduction to us in South Africa as he is an old Witsie hailing from Emmarentia in Johannesburg. His work on the role of central sensitization and its implications have proven the basis for much of what we do in everyday practice and the article summarises most of what we, as treating practitioners, need to know. Orofacial pain is another topic that deserves much more attention and the review by Hargreaves will subject that seems to be making the headlines is post-operative pain as it is well known that this type of pain is poorly managed. I shall try and publish as many of the review series as possible whenever the Biennial Review is receive are as practical as the Review Series and it is after all the practice of pain management that is of great concern to all of us no matter what our primary discipline may be. I wish you all well over the Festive Season and trust that you all have a good rest during the holiday season. Next year is going to be a busy one as there are many good local meetings supported by painsa including the Wits Pain Symposium, the Pretoria Pain Meeting and of course the highlight being the PAINSA Annual Congress being organized by Dr Sean Chetty and his Committee. This congress Dr. Milton Raff BSc (WITS), MBChB (Pret), FFA (SA) All correspondence to the editor should be addressed to: raffs@iafrica.com 1

4 EDITOR Dr. M Raff BSc (WITS), MBChB (Pret), FFA (SA) EDITORIAL BOARD Prof. H Meyer MBChB(Pret) MPraxMed(Pret) MFGP(SA) Prof. C L Odendaal Prof. D Mitchell BSc Hons, MSc, PhD (all University of the Witwatersrand) Dr. S Baumann BA. Mb.Ch.B.(U.C.T.), P.G.C.E.(University College of Wales), M.R.C.Psych.(London), F.C.Psych (S.A.) Mrs. P Berger BSc Physio (Wits), Acup (SA) Dr. E Frohlich MD(Tel-Aviv), DA(SA), FCA(SA), Master (Med) Pain Management (Syd) Central sensitization: Implications for the diagnosis and treatment of pain Clifford J. Woolf Orofacial pain Kenneth M. Hargreaves Pathophysiology of postoperative pain Timothy J. Brennan This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, reprinting reuse of illustrations, broad-casting, reproduction of CD- or in any other way, and storage in data banks. The use of registered names trademarks etc. in this publication does not imply, even in the absence of are exempt for the relevant laws and regulations and therefore free for general use. Product liability: the publishers cannot guarantee the accuracy of any information about the publication of medications contained in this publication. In every individual case, the user must check such information by consulting the relevant literature. PUBLISHER / MEDSPEC PUBLISHING Reni Rouncivell, Tel: (012) Fax: , Cell: , reni@medspec.co.za, Private Bag X1036, Lyttelton, South Africa 0140 ADVERTISING & RATES Sue-Anne Smook, Tel: (012) Fax: , Cell: , sueanne@medspec.co.za, Private Bag X1036, Lyttelton, South Africa 0140 Lelani Adendorff, Tel: (012) Fax: , Cell: , lelani@medspec.co.za, Private Bag X1036, Lyttelton, South Africa 0140 SUBSCRIPTIONS & ACCOUNTS Elizabeth Versteeg, Tel: , accounts@medspec.co.za FOR ADDRESS CHANGES PLEASE CONTACT: PAIN SA MEMBERS: YVONNE PYNE-JAMES, painsa@uiplay.com / NON PAIN SA MEMBERS: LINDA VANDERBERG, linda@promailhealthcare.co.za PHYSIOTHERAPY GROUP: BEVERLEY BOLTON, abcdbolt@telkomsa.net / TRADE: CALLY LAMPRECT, cally@medspec.co.za 2

5

6

7 PAIN Ò 152 (2011) S2 S15 Review Central sensitization: Implications for the diagnosis and treatment of pain Clifford J. Woolf Program in Neurobiology and FM Kirby Neurobiology Center, Children s Hospital Boston, Department of Neurobiology, Harvard Medical School, Boston, MA, USA Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article. article info abstract Article history: Received 9 August 2010 Received in revised form 24 September 2010 Accepted 24 September 2010 Nociceptor inputs can trigger a prolonged but reversible increase in the excitability and synaptic efficacy of neurons in central nociceptive pathways, the phenomenon of central sensitization. Central sensitization manifests as pain hypersensitivity, particularly dynamic tactile allodynia, secondary punctate or pressure hyperalgesia, aftersensations, and enhanced temporal summation. It can be readily and rapidly elicited in human volunteers by diverse experimental noxious conditioning stimuli to skin, muscles or viscera, and in addition to producing pain hypersensitivity, results in secondary changes in brain activity that can be detected by electrophysiological or imaging techniques. Studies in clinical cohorts reveal changes in pain sensitivity that have been interpreted as revealing an important contribution of central sensitization to the pain phenotype in patients with fibromyalgia, osteoarthritis, musculoskeletal disorders with generalized pain hypersensitivity, headache, temporomandibular joint disorders, dental pain, neuropathic pain, visceral pain hypersensitivity disorders and post-surgical pain. The comorbidity of those pain hypersensitivity syndromes that present in the absence of inflammation or a neural lesion, their similar pattern of clinical presentation and response to centrally acting analgesics, may reflect a commonality of central sensitization to their pathophysiology. An important question that still needs to be determined is whether there are individuals with a higher inherited propensity for developing central sensitization than others, and if so, whether this conveys an increased risk in both developing conditions with pain hypersensitivity, and their chronification. Diagnostic criteria to establish the presence of central sensitization in patients will greatly assist the phenotyping of patients for choosing treatments that produce analgesia by normalizing hyperexcitable central neural activity. We have certainly come a long way since the first discovery of activity-dependent synaptic plasticity in the spinal cord and the revelation that it occurs and produces pain hypersensitivity in patients. Nevertheless, discovering the genetic and environmental contributors to and objective biomarkers of central sensitization will be highly beneficial, as will additional treatment options to prevent or reduce this prevalent and promiscuous form of pain plasticity. Ó 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved. 1. Introduction In 1983 I published a study indicating that many features of the pain hypersensitivity accompanying peripheral tissue injury or inflammation were the direct result of an augmentation of sensory signaling in the central nervous system [255]. A central amplification during angina pectoris had been postulated exactly 100 years before by W. Allen Sturge MD, who in an 1883 paper in Brain envisaged a possible central nervous system commotion... passed up from below that somehow contributed to the clinical features of ischemic cardiac pain. However, the importance of this clinical insight lay largely dormant for a century, except for one human volunteer study on secondary hyperalgesia that was recognized by the authors as suggestive of a possible central contribution to the spread of pain sensitivity [101]. What I found in a pre-clinical address: clifford.woolf@childrens.harvard.edu study on stimulus response relations in the spinal cord was that the afferent activity induced by peripheral injury triggered a long-lasting increase in the excitability of spinal cord neurons, profoundly changing the gain of the somatosensory system [255]. This central facilitation manifested as a reduction in threshold (allodynia), an increase in responsiveness and prolonged aftereffects to noxious stimuli (hyperalgesia), and a receptive field expansion that enabled input from non-injured tissue to produce pain (secondary hyperalgesia) [51, ,268,273]. I have recently reviewed the circumstances surrounding the discovery of the activity-dependent synaptic plasticity in the spinal cord that generates post-injury pain hypersensitivity [259], and that became termed central sensitization [272], as well as the current state of understanding of the cellular and molecular mechanisms responsible for this form of neuronal plasticity [147]. What I would like to specifically address in this review are the clinical implications of the phenomenon. What has central sensitization taught us about the nature and mechanisms of pain in patients, /$36.00 Ó 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved. doi: /j.pain

8 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S3 and what are the implications of central sensitization for pain diagnosis and therapy? Before doing this though, it is important first to understand exactly what central sensitization represents, how it has changed our general understanding of pain mechanisms, as well as reviewing the substantial data on central sensitization derived from studies on experimental pain in human volunteers. 2. What is central sensitization? Prior to the discovery of central sensitization, the prevailing view on pain processing in the central nervous system was of a largely passive neural relay that conveyed by encoded action potentials, information on the onset, duration, intensity, location and quality of peripheral noxious stimuli, much like a telephone wire, from one site to another. More specifically, the CNS pathway was seen to constitute particular anatomical connections in the spinal cord, brain stem, thalamus and cortex (the pain pathway ), linking the sensory inflow generated in high threshold primary afferents with those parts of the cortex that leads to the conscious awareness of painful sensations. The spinal gate control theory by Melzack and Wall in 1965 had highlighted that this sensory relay system could be modulated in the spinal cord by inhibitory controls [163], and considerable progress had been made by the early 1980 s in identifying such inhibitory circuits [18]. Indeed this, together with the discovery of enkephalins and endorphins [98,109], diffuse noxious inhibitory controls [150], transcutaneous nerve stimulation [224], and the rediscovery of acupuncture [25], generated a much greater emphasis at that time on endogenous inhibitory controls than on those factors that might increase excitation, and thereby produce pain hypersensitivity. However, there was one exception, which was related to the discovery of peripheral sensitization in the 1970 s [178]. Work by Iggo [28,112] and Perl [20,33,177] had identified specific high threshold sensory neurons tuned to respond only to noxious stimuli, hence their name nociceptors [265], a term first coined by Sherrington based on his studies on noxious stimulus evoked flexion reflexes. Furthermore, first Perl and then others showed that nociceptor peripheral terminals could become sensitized after injury, reducing their threshold, mainly to heat stimuli, and only within the site of injury where the terminal was exposed to inflammatory modulators, the zone of primary hyperalgesia [23,41,138,146,178]. While this phenomenon is clearly a very important contributor to inflammatory pain hypersensitivity [22], it cannot account for dynamic tactile allodynia, the temporal summation of pain, or secondary hyperalgesia. Some other explanation was needed as the neurobiological basis for these symptoms, which turned out to be increased synaptic function triggered within the CNS by nociceptive inputs [257,237,268]. The realization that synapses were subject to a form of usedependent plasticity that could increase their strength or efficacy had steadily gained ground by the early 1980 s. The phenomenon had first been described in the CNS as short lasting a post-tetanic potentiation of mono synaptic IA synaptic input to motor neurons by Lloyd in 1949 [155], one that could spread to other synapses on motor neurons [21]. This was followed by the discovery of windup in dorsal horn neurons by Mendell and Wall in 1965 [164], where repeated low frequency stimulation of a nerve at constant C-fiber strength was found to elicit a progressive increase in action potential firing over the course of the stimulus. A transformative breakthrough was the first description of long term potentiation (LTP) in the hippocampus by Bliss and Lomo in 1973, where a brief high frequency coincident input produced a persistent increase in synaptic efficacy, opening the door for an extensive and still ongoing study into the molecular mechanisms of synaptic plasticity. LTP was first recorded in the spinal cord in 1993 [182], where it represents a particular component of the general phenomenon of central sensitization [113,114,122]. In 1976 Kandel and colleagues described a sensitization of the gill withdrawal reflex in the sea snail Aplysia, which was associated with a facilitation of the synapse between sensory and motor neurons [29]. However, these data were interpreted as reflecting memory and learning rather than an invertebrate model of pain hypersensitivity, although of course the two phenomena converge in this, and in other model systems, although there are differences too [122,274]. What I found in my original study by 1983 and subsequent preclinical studies with colleagues at University College London was that a brief (10 20 s), low frequency (1 10 Hz) burst of action potentials into the CNS generated by electrical stimulation or natural activation of nociceptors increased synaptic efficacy in nociceptive neurons in the dorsal horn of the spinal cord and this lasted for tens of minutes after the end of the conditioning stimulus [50,51,230,244,245,255,256,263,264,267,272,273]. This phenomenon differed from windup, which represented a progressively increasing output during the course of a train of identical stimuli (technically called homosynaptic potentiation); central sensitization was concerned instead with the facilitation that manifested after the end of the conditioning stimuli, and that once triggered remained autonomous for some time, or only required a very low level of nociceptor input to sustain it. Furthermore, central sensitization represented a condition where input in one set of nociceptor sensory fibers (the conditioning input) amplified subsequent responses to other non-stimulated non-nociceptor or nociceptor fibers (the test input; this form of facilitation is termed heterosynaptic potentiation to distinguish it from homosynaptic potentiation where the test and conditioning input are the same) [231]. The classic form of LTP in the hippocampus is homosynaptic with changes in efficacy restricted to activated synapses, a convergent plasticity, and while this is a feature of some aspects of central sensitization [190], most of its clinically relevant attributes relate to its divergent heterosynaptic components [147]. The underlying neurobiological basis for central sensitization is that for most central circuits, the receptive field properties of neurons defined by the firing of action potentials is only the tip of the iceberg. Most of the synaptic input to neurons is subthreshold [262,263], acting subliminally either because synaptic input is too weak or membrane excitability is restrained by inhibitory inputs. Increasing synaptic strength by a presynaptic increase in an excitatory transmitter release or in the post synaptic response to the transmitter [46,100,129,130,133,151,152,154,227,231,247,264,271] or by reducing inhibition [12,103,168,180,165,208,226] or increasing membrane excitability can recruit these normal subthreshold inputs to suprathreshold action potentials, producing profound changes in functional properties [270]. More recently it has become appreciated that in addition to activity-dependent synaptic plasticity, changes in microglia, astrocytes, gap junctions, membrane excitability and gene transcription all can contribute to the maintenance of central sensitization [43,44,47,48,88,104,186,189, 205,234]. Figs. 1 and 2 summarize sensory processing under normal circumstances and the changes that result from induction of central sensitization. An important implication of these early basic science studies was the possibility that the pain we experience might not necessarily reflect the presence of a peripheral noxious stimulus. We learn from our everyday experience interfacing with the external environment to interpret pain as reflecting the presence of a peripheral damaging stimulus, and indeed this is critical to its protective function. Central sensitization introduces another dimension, one where the CNS can change, distort or amplify pain, increasing its degree, duration, and spatial extent in a manner that no longer directly reflects the specific qualities of peripheral noxious stimuli, but rather the particular functional states of circuits in the CNS. With the discovery of central sensitization, pain 6

9

10 S4 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 Fig. 1. Normal sensation. The somatosensory system is organized such that the highly specialized primary sensory neurons that encode low intensity stimuli only activate those central pathways that lead to innocuous sensations, while high intensity stimuli that activate nociceptors only activate the central pathways that lead to pain and the two parallel pathways do not functionally intersect. This is mediated by the strong synaptic inputs between the particular sensory inputs and pathways and inhibitory neurons that focus activity to these dedicated circuits. Fig. 2. Central sensitization. With the induction of central sensitization in somatosensory pathways with increases in synaptic efficacy and reductions in inhibition, a central amplification occurs enhancing the pain response to noxious stimuli in amplitude, duration and spatial extent, while the strengthening of normally ineffective synapses recruits subliminal inputs such that inputs in low threshold sensory inputs can now activate the pain circuit. The two parallel sensory pathways converge. conceptually at least had become centralized instead of being exclusively peripherally driven. In this sense central sensitization represents an uncoupling of the clear stimulus response relationship that defines nociceptive pain. Nociceptive pain reflects the perception of noxious stimuli. In the absence of such potentially damaging stimuli there is no nociceptive pain. However, after the discovery of central sensitization it became clear that a noxious stimulus while sufficient was not necessary to produce pain. If the gain of neurons in the pain pathway in the CNS was increased, they could now begin to be activated by low threshold, innocuous inputs. In consequence pain could in these circumstances become the equivalent of an illusory perception, a sensation that has the exact quality of that evoked by a real noxious stimulus but which occurs in the absence of such an injurious stimulus. This does not mean that the pain is not real, just that it is not activated by noxious stimuli. Such pain can no longer be termed nociceptive, but rather reflects a state of induced pain hypersensitivity, with almost precisely the same symptom profile to that found in many clinical conditions. This raised the immediate obvious question, was central sensitization a contributor to clinical pain hypersensitivity? These notions were generally not very well received initially, particularly by physicians who believed that pain in the absence of pathology was simply due to individuals seeking work or insurance-related compensation, opioid drug seekers, and patients with psychiatric disturbances; i.e. malingerers, liars and hysterics. That a central amplification of pain might be a real neurobiological phenomenon, one that contributes to diverse clinical pain conditions, seemed to them to be unlikely, and most clinicians preferred to use loose diagnostic labels like psychosomatic or somatoform disorder to define pain conditions they did not understand. We can now 30 years later, based on data from many studies in human 8

11 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S5 volunteers and patients, address whether central sensitization, defined operationally as an amplification of neural signaling within the CNS that elicits pain hypersensitivity, is a real phenomenon or not, and can assess its relative contribution to inflammatory, neuropathic and dysfunctional pain disorders in patients [53,258]. 3. Central sensitization in human volunteers The first clear demonstration of central sensitization in human volunteers came from a psychophysical study by LaMotte and colleagues on the secondary cutaneous hyperalgesia that is elicited by intradermal capsaicin injection (which activates the TRPV1 receptor). They found intense localized pain lasting minutes at the injection site, followed immediately by three zones of hyperalgesia; a small zone of heat hyperalgesia close to the injection site lasting 1 2 h, an intermediate zone of dynamic tactile allodynia spreading beyond the area of heat hyperalgesia and lasting several hours, and the largest zone to pinprick, way outside of the injection site, which remained present for up to 24 h [145]. The investigators then showed that the secondary mechanical hyperalgesia required sensory inflow to the CNS because local anesthesia prior to the capsaicin injection blocked it. In addition because the pain sensitivity crossed a tight band that prevented circulation in the skin, they concluded that it was not due to a local spread of the capsaicin or any peripheral inflammatory mediator. An even more direct demonstration that activity-dependent central sensitization was responsible for tactile allodynia and secondary hyperalgesia in humans came from a second study by La Motte, this time with Torebjork in 1992 [233]. They again used intradermal injection of capsaicin to induce an area of tactile allodynia that lasted for 2 h. Nerve block experiments revealed that while the capsaicin and heat pain was carried by C fibers, the mechanical allodynia was transferred to the CNS by low threshold myelinated fibers. The most elegant part of the study was their finding that electrical intraneural stimulation of single Ab mechanoreceptive fibers that elicited a non-painful tactile sensation before the capsaicin injection, began to produce pain if the fibers receptive field fell within the zone of secondary mechanical hyperalgesia. Lidocaine anesthesia of the cutaneous innervation territory of the stimulated fiber did not reverse the pain, showing that this was not peripheral in origin. They concluded that the pain evoked by stroking the skin area surrounding a painful intradermal injection of capsaicin is due to reversible changes in the central processing of mechanoreceptive input from myelinated fibres which normally evoke nonpainful tactile sensations. Another early study, this time by Koltzenburg and Torebjork, using mustard oil (which activates TRPA1) as the pain conditioning stimulus, together again with differential nerve blocks, confirmed that brush-evoked mechanical allodynia was mediated by low threshold Ab fibers that normally encode non-painful tactile sensations [140]. Unlike after capsaicin, however, the mustard oil evoked tactile allodynia required an ongoing low level input from C-nociceptors to sustain it, indicating that different sensory fibers may have different central actions, some short and others long lasting, and indeed further studies have shown differences in the duration of tactile allodynia after capsaicin and mustard oil [139], the significance of which was not appreciated a the time because it was not clear then that these irritants acted on quite different TRP receptors. That central sensitization could cause a spread of pain sensitivity across peripheral nerve territories, the neurological dogma for diagnosing a disease of the central rather than peripheral nervous system was shown by Max and colleagues using the intradermal capsaicin model in volunteers together with radial or ulnar nerve blocks to clearly identify individual nerve territory [192]. Complementing this, a study comparing skin hyperaemia induced by a skin burn injury found that the skin blood flow changes induced by the injury had disappeared by the time secondary mechanical hyperalgesia peaked, and the two were not correlated in time or space, supporting the conclusion that peripheral mechanisms do not contribute to secondary hyperalgesia [198]. Perhaps even more dramatic, was the relatively recent demonstration that intradermal capsaicin induces contralateral hyperalgesia and allodynia that are delayed in their manifestation and reduced in extent compared to the ipsilateral secondary hyperalgesia, but present in a majority of subjects [206], a form perhaps of tertiary hyperalgesia that cannot be peripheral in origin. What pain sensitivity we feel then, can be determined by the state of excitability of neurons in the CNS. Central amplification of Ad nociceptor fiber test input following a C-fiber conditioning input was shown to contribute to pinprick/ punctate secondary hyperalgesia, again using the intradermal capsaicin model [279], underscoring the different identity of the afferent signals that elicit central sensitization as a conditioning stimulus (C-fibers) from those that elicit allodynia (Fb) or hyperalgesia (Ad), a further clear manifestation of heterosynaptic facilitation. In a similar vein, another study found that pin prick hyperalgesia induced in response to intradermal capsaicin was actually mediated by capsaicin-insensitive afferents, showing that the test and conditioning inputs in this setting are quite different [87], while the secondary hyperalgesia elicited by intradermal capsaicin was shown by yet other investigators, to be restricted to mechanical stimuli, with no correlation between the magnitude of capsaicin evoked pain and the extent of punctate or tactile secondary hyperalgesia [237]. Furthermore, temporal summation to pin prick in the zone of capsaicin injection (as model of homosynaptic facilitation/windup) was mechanistically independent of the development of secondary hyperalgesia, because while the gain of the stimulus response relationship in the zone of secondary was increased that of the windup was not changed, even though the actual pain was enhanced [158]. A similar conclusion was made after a study where repeated intradermal capsaicin injections were reported to produce a progressively diminishing pain, presumably due to desensitization, while the allodynia and punctate hyperalgesia continued to increase [254]. Two more recent studies using high frequency stimulation as the conditioning input to mimic conditions that elicit LTP found that while changes in the conditioned site (homotopic site) do occur, they are accompanied by a development of pain hypersensitivity in the adjacent non-stimulated heterotopic site (reduction in threshold, pain evoked by light tactile stimuli, and exaggerated response to suprathreshold pinprick stimuli [136,240], and both sets of investigators concluded that heterosynaptic facilitation predominates in this model of central sensitization, exactly as it does for the low frequency conditioning inputs that mimic the natural firing range of nociceptors. Generalizing, it seems clear that heterosynaptic changes are a major feature of the presentation of central sensitization. Apart from changes in subjective pain measures, the consequences of central sensitization can also be detected using objective biomarkers. These include long-term changes in nociceptive withdrawal reflexes [24] and increases in cortical event related potential amplitudes [240]. Magnetic source imaging reveals an increase in the excitability of neurons in the somatosensory cortex evoked by low threshold Ab stimulation within the capsaicin-induced zone of secondary hyperalgesia [17], while magnetoencephalography detects changes in the patterns of cerebral processing [159] and functional MRI, and changes in BOLD signals in the cortex, both during secondary hyperalgesia [16]. Another MRI study found changes in the brainstem that are apparently specific to central sensitization, in addition to the changes in the primary somatosensory cortex that are related to the intensity of pain [153]. While most studies have looked at the effects of skin conditioning stimuli on skin pain sensitivity, experimental muscle pain produced by hypertonic saline injections produces long lasting 9

12 S6 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 changes in thermal sensitivity in the area of referred pain [203], while sustained nociceptive stimulation of myofascial trigger points induces a wide spread central sensitization [273,275]. Interestingly, in pre-clinical models, muscle and joint conditioning afferents have a longer lasting action in producing central sensitization than those from skin [244]. A reverse approach has shown that cutaneous capsaicin increases myofascial trigger point pressure sensitivity in segmentally related muscles [211]. Conditioning nociceptive stimuli originating in viscera, such as exposure of the lower esophagus to acid, also induces central sensitization, leading to viscerovisceral (pain hypersensitivity in the upper esophagus) and viscerosomatic hypersensitivity (allodynia on the chest wall) [193] that can be captured by esophageal evoked potentials [194], and is associated with increased temporal summation [196]. A recent study has replicated this esophageal model of central sensitization using acid and capsaicin infusions, showing also thermal and mechanical pain hypersensitivity in the rectum after the esophageal stimulation [27], indicating how widespread the effects of central sensitization are in the gastro-intestinal tract. These changes may be mechanistically related to widespread clinical pain syndromes [95]. One emerging area of considerable interest is the utility of experimental central sensitization in human volunteers to test efficacy in centrally acting drugs. Drugs with efficacy in pre-clinical models, such as NMDA receptor antagonists [271] can be tested in Phase 1b human proof of principle studies [212]. Ketamine inhibits central temporal summation [8] and secondary mechanical hyperalgesia [142] evoked by repetitive nociceptive electrical stimulation in humans as well as primary and secondary hyperalgesia after an experimental burn injury [116], visceral conditioning inputs [251,253] and topical [6] or intradermal [204] capsaicin, but not A delta mediated nociceptive pain [181]. Ketamine s action on experimental pain can be detected by fmri [210]. Similar activity is found for i.v. dextromethophan [115]. Collectively these data strongly support a role for the NMDA receptor in acute activitydependent central sensitization [147]. However, the trials also indicate the lack of therapeutic index between reducing central sensitization and inducing psychotomimetic side effects. Another class of drugs that has been extensively studied in human experimental models of central sensitization is the gabapentanoids. Oral gabapentin at doses similar to that used for chronic neuropathic pain when given to human volunteers reduced tactile allodynia and decreased mechanical secondary hyperalgesia elicited by intradermal capsaicin [92]. Even single administration of gabapentin had an antihyperalgesic effect on capsaicin-induced secondary hyperalgesia and reduced fmri signatures of central sensitization [110]. In another study gabapentin, interestingly reduced cutaneous evoked central sensitization but not muscle pain [201]. Two studies have looked at pregabalin s efficacy in experimental human central sensitization, one evoked by electrical stimuli [49] and the other by intradermal capsaicin [246]. Both of these double blind studies demonstrated efficacy for pregabalin in terms of experimental tactile allodynia and secondary hyperalgesia. These data suggest that a major component of gabapentin or pregabalin s mechanism of action is a reduction of central sensitization [238]. Many other centrally acting drugs with analgesic efficacy in patients reduce central sensitization preclinically, including duloxetine, milnacipran and lamotrigene [15,118,170] but have not been tested for this action in humans. Drugs that have failed to show efficacy in human studies of activity-dependent central sensitization are NK1 receptor antagonists [252] [49] and COX-2 inhibitors [35,49,250]. A COX-2 inhibitor does have efficacy though if the central sensitization is triggered by peripheral inflammation [225], as predicted by pre-clinical models [189]. Interestingly, while gender has been described as important for differences in nociceptive pain sensitivity, a study on the secondary hyperalgesia induced by heat and capsaicin did not reveal a gender difference [119]. Nevertheless, recent data show that pain sensitivity including secondary hyperalgesia and brush evoked allodynia is heritable, with an estimated 50% genetic contribution to the pain variance [172]. The genetic polymorphisms involved in the differential susceptibility to secondary hyperalgesia have not been comprehensively investigated, although some candidates are beginning to be identified in studies of experimental central sensitization [228]. This is an area that requires major research. The following conclusions can be made from this survey of the published studies of experimental pain hypersensitivity in human volunteers. Central sensitization is a robust phenomenon, readily induced in human volunteers in response to diverse ways of activating nociceptors (electrical stimulation, capsaicin, mustard oil, acid, heat burn, UV burn, hypertonic saline). Generally this activity-dependent plasticity manifests immediately, but its effects persist for many hours beyond the inducing conditioning stimulus, eventually returning, however, back to baseline, indicating its usual full reversibility. The phenomenon can be elicited by conditioning skin, muscle or visceral organs, and typically presents as dynamic tactile allodynia and punctate hyperalgesia but also enhanced pressure, and in some cases, thermal sensitivity, spreading from the conditioning site to neighboring non-stimulated sites, and even to very remote regions. Although there is a homosynaptic (homotopic) aspect to the phenomenon, its major manifestation is heterosynaptic (heterotopic), and for this reason and its reversibility, it is perhaps inaccurate to equate central sensitization with the LTP like phenomena in the cortex that are specifically associated with long term memory. Because central sensitization can be induced in almost all subjects and detected using subjective and objective outcome measures and is sensitive to pharmacological interventions, it is a useful tool for determining the activity of drugs on centrally driven pain hypersensitivity. Globally, the data obtained in human volunteer studies demonstrate that induction of use-dependent central facilitation in nociceptive central pathways increases pain sensitivity and may, therefore, contribute to clinical pain syndromes. Experimental studies in human volunteers are necessarily restricted to use non-injurious conditioning inputs, and therefore are limited to studying only the activity-dependent components of pain hypersensitivity elicited by sensory inputs, and not those transcription-dependent and structural changes that manifest after inflammation or nerve injury, which may have different mechanisms, time courses and presentations [53,97,121,123,160,171, 189,229,242,261,269]. The limited experience with more severe human experimental injury indicates that central sensitization also contributes to the late hyperalgesia present in this model [58,176]. 4. Central sensitization and the clinical pain phenotype What features of the clinical phenotype may be contributed to, or generated exclusively by central sensitization? While the human experimental studies reviewed above indicate that if a patient has dynamic tactile allodynia, secondary punctuate/pressure hyperalgesia, temporal summation and sensory aftereffects, central sensitization may well be involved. Any sensory experience greater in amplitude, duration and spatial extent than that would be expected from a defined peripheral input under normal circumstances qualifies as potentially reflecting a central amplification due to increased excitation or reduced inhibition. These changes could include a reduction in threshold, exaggerated response to a noxious stimulus, pain after the end of a stimulus, and a spread of sensitivity to normal tissue. However, because we cannot directly measure sensory inflow, and because peripheral changes can contribute to sensory amplification, as with peripheral sensitization, pain hypersensitivity by itself is not enough to make an 10

13 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S7 irrefutable diagnosis of central sensitization. A further complication is that because peripheral input commonly is the trigger of central sensitization, a reduction in pain sensitivity produced by targeting a peripheral trigger with a local anesthetic does not exclude central amplification, but may rather indicate a role of peripheral input in maintaining it [140]. Nevertheless, there are some features of patient s symptoms which are more likely to indicate central rather than peripheral contribution to pain hypersensitivity. These include pain mediated by low threshold Ab fibers (determined by nerve block or electrical stimulation), a spread of pain sensitivity to areas with no demonstrable pathology, aftersensations, enhances temporal summation, and the maintenance of pain by low frequency stimuli that normally do not evoke any ongoing pain. To assess how central sensitization may present in patients, we need a detailed phenotyping of different patient cohorts to capture exactly what changes in sensitivity occur, where and when [9,11,55,86,93,188,197]. Ideally this should be combined with objective measures of central activity, such as fmri, so that clear diagnostic criteria for determining the presence of central sensitization in patients can be established. The utility of diagnostic criteria for the presence of central sensitization would not only be insight into the pathophysiological mechanisms responsible for producing pain but more so in defining potential treatment strategies. If a particular patient s pain is primarily the result of abnormal activity in nociceptors, as in patients with primary erythromelalgia [74], the optimal therapy required is likely to be different from a patient whose tactile allodynia and secondary hyperalgesia are entirely maintained by central sensitization due to changes in synaptic efficacy in the spinal cord. This is the rationale for a mechanism-based approach to the diagnosis and treatment of pain [258,266]. Indeed response to a trial treatment, such as to the NMDA receptor antagonist ketamine, can itself be a potential diagnostic for the presence central sensitization. 5. To which clinical syndromes does central sensitization contribute? Given the caveats about the lack of absolute diagnostic criteria for identifying the presence of central sensitization in patients, a fairly large number of studies have nevertheless putatively identified this phenomenon as contributing to patients pain phenotype. I will briefly review these, based on disease Rheumatoid arthritis (RA) Patients with RA, the prototypic inflammatory joint disease, have extra-articular tenderness which is correlated with the extent of joint disease [141] but whether this is the result of peripheral or central sensitization has not been studied. A study on juvenile chronic arthritis reported enhanced sensitivity to noxious stimuli both at joints and in remote areas in patients with and without active disease, suggesting the possibility that the disease when active sets up a state of autonomous central sensitization [107] Osteoarthritis (OA) This degenerative joint disease with characteristic destruction of cartilage and alteration in bone is a very common cause of chronic pain, particularly in the elderly. The degree of pain does not always correlate with the extent of joint damage or presence of active inflammation raising the possibility that there may be a central component to the pain [26]. Supporting this is the enhanced degree and duration of pain and secondary hyperalgesia evoked by intramuscular injection of hypertonic saline in patients with OA compared to controls [13]. Patients with high pre-operative pain and a low pain threshold have a higher risk of persistent pain after total knee replacement for OA, which was interpreted as reflecting central sensitization [157]. Another study on 62 patients showed that pain of central neural origin (widespread reduced pressure pain thresholds) negatively impacted on knee functional capacity [117]. OA patients have a lower pain threshold and have punctate hyperalgesia in areas of referred pain, which is associated with greater activation in the brainstem as detected by fmri, representing a possible biomarker for central changes [99]. The centrally acting amine uptake inhibitor duloxetine which reduces central sensitization in pre-clinical models [15,124] significantly reduced pain more than placebo in an RCT in 231 patients with knee OA pain [45], indicating that drugs that target central sensitization are efficacious in this patient population. In a recent phenotyping study in 48 patients with painful knee OA and 24 age matched controls, the patients had reduced pressure pain thresholds both at the joint and in remote areas, and increased temporal summation. While the degree of sensitization correlated with the pain, it did not correlate with radiological findings, leading to the conclusion that central sensitization is an important contributor to knee OA pain [7]. Collectively, these data intriguingly suggest that the pain of OA, a peripheral pathology, has an important central component, and this is clearly deserving more study to understand its extent, mechanism and therapeutic implications Temporomandibular disorders (TMD) Unlike OA, the pathophysiology of this syndrome is much less well understood. However, TMD has been found to be associated with an increase in generalized pain sensitivity after isometric contraction of the orofacial muscles [166], and widespread bilateral mechanical [78] and thermal [175] pain sensitivity are reported in women with myofascial TMD compared to age matched controls, which was interpreted as suggesting widespread central sensitization. In addition, a greater referred pain is elicited from the more frequent trigger points that are found in these patients, than in controls [77]. As for other types of facial pain, mechanical allodynia is a major feature of periradicular inflammation (periradicular periodontitis) with reduced threshold also in contralateral non inflamed teeth, reflecting central sensitization [132]. After a third molar extraction evidence for central sensitization could be detected for at least a week (enhanced response to repetitive intraoral pinprick and electrical stimulation, aftersensations and extraoral hyperalgesia) [126] Fibromyalgia (FM) One of the first suggestions that fibromyalgia patients may have generalized central sensitization came from a psychophysical study that identified widespread reduction in thermal and mechanical pain thresholds, as well as greater cerebral laser evoked potentials [90], a finding replicated soon after [156]. Another early small study using ketamine, showed an NMDA-dependent component to fibromyalgia and suggested that tender points may represent secondary hyperalgesia due to central sensitization [209]. Supporting this, Arendt-Nielson and colleagues found in small study that fibromyalgia patients had lower pressure thresholds and increased temporal summation to muscle stimulation, and that intramuscular hypertonic saline injections provoked a longer lasting and more widespread pain. In a related study, they found that the referred pain, temporal summation, muscular hyperalgesia and muscle pain in fibromyalgia patients were all attenuated by ketamine [96]. In 2001, Staud and Price begun a series of studies on fibromyalgia, first showing temporal summation and after sensations of the pain elicited by repetitive cutaneous thermal stimuli and repetitive mechanical stimuli to muscles 11

14 S8 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 [221]. In a second study they found that temporal summation occurred at substantially lower forces and at a lower frequency of stimulation in fibromyalgia patients than in control subjects, and that painful after sensations were greater in amplitude and more prolonged [215]. The enhanced experimental pain in fibromyalgia patients was shown to contribute to the variance of the clinical pain [220]. These investigators then showed that the maintenance of experimentally induced pain in fibromyalgia patients requires significantly less frequent stimulation than in normal controls, and concluded that this heightened sensitivity to very low frequency inputs contributes to the persistent pain in these patients [218]. A later study showed that the temporal summation of pain and its maintenance was widespread, and could be equally elicited from hands or feet, leading to the conclusion that central sensitization in these patients was generalized across the neuraxis [219]. In an fmri study they then found a stimulus and frequency dependent activation in several brain regions in fibromyalgia patients and controls, including ipsilateral and contralateral thalamus, medial thalamus, S1, bilateral S2, mid- and posterior insula, rostral and mid-anterior cingulate cortex. The stimulus temperatures necessary to evoke equivalent levels of brain activity were, however, significantly less in fibromyalgia patients, suggesting that the enhanced neural mechanisms in fibromyalgia are not the result of selective enhancement at cortical levels [216]. The Staud and Price group then designed experiments to see if peripheral sensitization may contribute to the enhanced temporal summation of thermal pain in fibromyalgia patients and concluded that it does not, based on thermal thresholds [214]. Recently they have found using local anesthetic injections though, that peripheral input from muscle appears to be important in maintaining central sensitization in FM patients [217]. This would mean that fibromyalgia may have both peripheral and central contributions, whose extent may vary from patient to patient. Certainly muscle afferents seem to have a potent capacity in pre-clinical [244] and experimental human studies [275] to induce central sensitization. A quantitative sensory testing study in 85 fibromyalgia patients and 40 matched controls found that the patients had altered heat and cold thresholds and a reduced tolerance for pain, as well as a reduced nociceptive reflex threshold, a measure of central excitability [65]. The latter finding was sufficiently different from controls that the authors suggest it could be used as a diagnostic measure of central sensitization, identifying patients for whom centrally acting drugs may be particularly beneficial. Other studies have confirmed the increased generalized sensitivity in FM patients to pressure and thermal stimuli [94,173,179] and to electrical stimulation of skin and muscle, with enhanced cortical evoked potentials [66]. The data overall seem to support a major role for central sensitization in the generation of the symptoms of FM, and the success of centrally acting treatments, such as pregabalin or duloxetine in treating these conditions, may reflect a reduction in central sensitization in these patients Miscellaneous musculoskeletal disorders Chronic neck pain resulting from whiplash is associated with lowered pain thresholds in uninjured tissue [57,222]. Injection of local anesthetic into myofascial trigger points in these patients results in an immediate increase in range of motion and elevation in pressure pain thresholds, which was felt to reflect dynamic maintenance of central sensitization by afferent triggers [85]. Patients with shoulder impingement syndrome also show widespread muscle sensitivity and an increased number of trigger points [105]. A widespread (bilateral) mechanical pain hypersensitivity is observed in patients with unilateral epicondylalgia (tennis elbow) interpreted as indicating central sensitization, possibly induced by a peripheral trigger [75]. Similar generalized deep tissue hyperalgesia can also be demonstrated in patients with chronic radiating low back pain with intervertebral disc herniation [173]. Collectively these data indicate that diverse musculoskeletal disorders are characterized by a spread of pain sensitivity to deep uninjured tissue and that low level peripheral inputs may maintain this Headache The first intimation that headaches have an important component mediated by central sensitization came from a study of spontaneous tension-type headaches which found that even in the absence of headache pericranial muscle tenderness was increased in patients compared to control subjects. During headache, muscle tenderness increased and thermal pain threshold decreased in the temporal region, but remained normal in the hand which was interpreted as an indication that segmental central sensitization contributed to pain in frequent sufferers of tension-type headache [120]. This was then followed by the observation by Bernstein and colleagues that cutaneous allodynia developed in 79% of patients during migraine attacks in, and sometimes beyond the area of referred pain [36,37]. This finding has been repeated in several studies since then [52,161,135,207]. While cephalic and extracephalic allodynia are well described, spontaneous body pain and allodynia have also been reported as preceding migraine attacks [56]. Laser evoked cutaneous pain thresholds are reduced during migraine attacks and cortical evoked potentials increased [62]. No change in heat pain thresholds are found in chronic tension-type headache, but there is pericranial tenderness [63,80] and hyperalgesia of neck shoulder muscles [81]. Nociceptive input from muscles has been suggested to contribute to the induction of central sensitization in tension-type headache [79], much as has been suggested for FM. In patients with cluster headaches the nociceptive flexion reflex threshold is reduced on the symptomatic side [191]. In a population study on primary headaches in 523 patients, evidence for pain hypersensitivity was found in those with tension type pain, with a greater disturbance in individuals with chronic or more frequent headaches, implying that central sensitization may contribute to the chronification of headache [30], something that is supported by epidemiological data [31]. In a longitudinal prospective study on whether increased pain sensitivity is a cause or an effect, a study in 100 individuals found that subjects had normal thresholds prior to the development of headache, but this decreased in those who then developed chronic tension-type headache, suggesting that the pain hypersensitivity is a consequence of frequent tension-type headaches, and not a predictor or risk factor [32], a finding interpreted as a showing that central sensitization plays a role in the chronification of tension-type headaches. Interestingly, a study in patients with either chronic migraine and chronic tension-type headache found in both cohorts reduced threshold for pressure, pinprick, blink, and the nociceptive flexion reflex, as well as higher windup ratios [83], possibly reflecting a common role for central sensitization in the chronification of different types of headache Neuropathic pain The first demonstration of a likely contribution of central sensitization to neuropathic pain came from a study by Campbell and colleagues, who showed that an ischemic conduction block of large myelinated fibers specifically reduced dynamic tactile allodynia [42], a finding that was soon replicated [140]. Since then careful phenotyping studies of conditions like carpal tunnel syndrome have revealed enhanced bilateral sensitivity and an extraterritorial spread of symptoms in patients with unilateral or single nerve entrapment, supporting a contribution of central sensitization [61,76,82,278]. Furthermore, ketamine reduces established periph- 12

15 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S9 eral neuropathic pain [125] and chronic phantom limb pain [73] indicating that ongoing activity- and NMDA receptor-dependent synaptic plasticity may contribute to maintain neuropathic pain. That tricyclic antidepressants, dual uptake inhibitors and calcium channel alpha(2)-delta ligands, all centrally acting drugs that normalize enhanced neural activity, are the current first line treatments for neuropathic pain [72], reinforces the importance of the central component of the pain and its suitability as a target for treatment Complex regional pain syndrome (CRPS) A prominent feature of chronic CRPS1 is tactile hyperesthesia and pressure hyperalgesia [241], which can be registered as enhanced S1 activation by a neuromagnetometer [243]. There is also thermal hyperalgesia in acute CRPS1 patients, which on the side ipsilateral to the diseased limb, may have a peripheral component due to ongoing aseptic inflammation, but the presence of contralateral hypersensitivity in the absence of any inflammatory changes points to an involvement of the CNS [108]. In a small randomized placebo controlled trial intravenous ketamine reduced CRPS pain [200] Post-surgical pain This is a very heterogenous group comprising acute postoperative pain and persistent pain of multiple causes, including surgically induced neuropathic pain [1,131]. In the acute phase, incisional pain is associated with a secondary punctate hyperalgesia that is ketamine sensitive [223], with no spread in thermal sensitivity [143] indicating induction of central sensitization. Considerable controversy exists over whether pre-emptive treatment targeting central sensitization is superior to postoperative treatment in treating either the acute postoperative pain or its transition to chronic pain [4,5,54,60,68,70,71,128,149,102,236,260]. Surprisingly, because of numerous technical problems related to the design, conduct and interpretation of such studies, this turns out to be a difficult issue to resolve [134,167]. This is not the place to review the full literature on pre-emptive analgesia, however my personal take on the available data is that there appears to be a small signal for pre- vs. postoperative analgesic treatment in some settings, but it is likely not generally clinically relevant. It seems clearly important though that patients have full analgesia established on recovery from a general anesthetic or adequate regional anesthesia during surgery, and this can be maintained until surgical healing is well advanced [19,14,277]. The treatment plan for controlling postoperative pain can potentially include drugs with action on central sensitization such as ketamine [184], pregabalin [34,162], gabapentin [202] and duloxetine [106], which in the limited number of trials currently available show some efficacy, but more RCT are required to assess their utility in treating acute postoperative pain or in reducing the risk of developing chronic pain [59] Visceral pain hypersensitivity syndromes Pain hypersensitivity is a feature of several common disorders of the gastro-intestinal tract including irritable bowel syndrome, non-cardiac chest pain and chronic pancreatitis that all appear to have a central sensitization component. A majority of IBS patients have both rectal and somatic hypersensitivity [249]. Repetitive sigmoid stimulation in patients with IBS induces rectal hyperalgesia and viscerosomatic referral [169]. Local rectal anesthesia reduces rectal and somatic pain in irritable bowel syndrome patients, supporting the possibility that visceral hyperalgesia and secondary cutaneous hyperalgesia in irritable bowel syndrome are the results of central sensitization dynamically maintained by input from the GIT. Patients with non-cardiac chest pain have esophageal hypersensitivity [195], with a reduced tolerance to repeated distension, increased size of referred pain and a greater propensity to show secondary hyperalgesia after acid infusion in their lower esophagus [69], all interpreted as reflecting the consequence of central sensitization. Chronic pancreatitis is associated with generalized deep pressure hyperalgesia [39,174] and patients display greater degree and spatial extent secondary hyperalgesia elicited by repetitive experimental stimulation, suggesting enhanced central sensitization [67] that is reduced by a thorascopic splanchnic denervation [38], which may reflect that visceral input from the pancreas maintains the central sensitization. In the urological tract, pain hypersensitivity is a feature of interstitial cystitis, chronic prostatitis, endometriosis, and vulvodynia, conditions whose pathophysiology and etiology are however, poorly understood. Although central sensitization has been hypothesized to contribute [137], not much data are available and few studies have been performed. Men with chronic prostatitis have though heightened pain sensitivity in the perineum [239,276], while women with vulvodynia have an enhanced post capsaicin allodynia and secondary hyperalgesia compared to controls [84] Co-morbidity of pain conditions characterized by pain hypersensitivity Pain can be defined as nociceptive when it is generated by noxious stimuli, inflammatory when produced by tissue injury and/or immune cell activation, and neuropathic, when it is due to a lesion of the nervous system. What about pain conditions though, where there is no noxious stimulus, inflammation or damage to the nervous system? There are several common syndromes that present with pain hypersensitivity but no clear etiological factor, i.e. considered unexplained and which might actually reflect not only peripheral pathology but also a primary dysfunction of the nervous system. These include fibromyalgia, tension-type headache, temporomandibular joint disease and irritable bowel syndrome, all of which may have a specific contribution to their phenotype by central sensitization, as detailed above. If a heightened sensitivity of the CNS or an increased propensity to develop central sensitization is a common feature of these syndromes, one would expect that there may be increased co-occurrence or comorbidity of the different conditions. It is also possible that an enhanced capacity to produce or maintain central sensitization is the primary defect in some of these syndromes. In a study on almost 4000 twins for comorbidity of chronic fatigue, low back pain, irritable bowel syndrome, chronic tension-type headache, temporomandibular joint disease, major depression, panic attacks and post-traumatic stress disorder, associations were found that far exceeded those expected by chance, and the conclusion was that these conditions share a common etiology [199]. Another large epidemiological study on 44,000 individuals including twins for comorbidity with chronic widespread pain found co-occurrence with chronic fatigue, joint pain, depressive symptoms, and irritable bowel syndrome, leading to the conclusion that associations between chronic widespread pain and its comorbidities may include genetic factors [127]. Yet another study on 2299 subjects for four unexplained syndromes; chronic wide spread pain, chronic orofacial pain, irritable bowel and chronic fatigue again found that the occurrence of multiple syndromes was greater than expected by chance [2]. These epidemiological findings strongly suggest that there may be a common mechanistic basis for these diverse conditions, and that may have a hereditary component. Smaller studies have found comorbidity between fibromyalgia and the following conditions: migraine in females but not males [111], primary headache [64], chronic fatigue symptom [89], 13

16 S10 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 systemic lupus erythematosus [213], irritable bowel syndrome [144], rheumatoid arthritis [183], the premenstrual syndrome [3], chronic urticaria [235] and cervical myofascial pain syndrome [40]. Comorbidity has been shown also for back pain and temporomandibular disorders [248], migraine and temporomandibular disorders [91], irritable bowel syndrome and functional dyspepsia, fibromyalgia and chronic pelvic pain [185], and finally between migraine and irritable bowel syndrome, chronic fatigue and fibromyalgia [232]. There is also an overlap between urological disorders like chronic pelvic pain, interstitial cystitis, painful bladder syndrome, chronic prostatitis and vulvodynia with fibromyalgia, chronic fatigue, temporomandibular disorders and irritable bowel syndrome [187], and more specifically between vulvodynia, fibromyalgia and irritable bowel syndrome [10]. The overwhelming conclusion from these diverse epidemiological studies is that chronic pain hypersensitivity in the absence of inflammation or nerve damage results in apparently phenotypically different syndromes depending on the tissue/organs affected. However, the overall similarity of the sensitivity changes may reflect a common contribution of central sensitization, and this may account for the unexpectedly high comorbid rate of the apparently different syndromes. To test if there are indeed central sensitization syndromes, we will need a clear set of diagnostic criteria and biomarkers for the phenomenon. If this hypothesis is correct, the implications may be that treatment strategies targeted at normalizing hyperexcitability in the CNS may have a shared efficacy for the different manifestations of the central sensitization syndrome. 6. Conclusions Clinical pain is not simply the consequence of a switching on of the pain system in the periphery by a particular pathology, but instead reflects to a substantial extent, the state of excitability of central nociceptive circuits. The induction of activity-dependent increases in synaptic function in these circuits triggered and maintained by dynamic nociceptor inputs, shifts the sensitivity of the pain system such that normally innocuous inputs can activate it and the perceptual responses to noxious inputs are exaggerated, prolonged and spread widely. These sensory changes represent the manifestation of central sensitization, and extensive experimental medicine and clinical investigations over the past twenty years have revealed it to be an important component of the pain hypersensitivity present many patients. While considerable progress has been made in teasing out the cellular and molecular mechanism responsible [148], much remains still to be learned, particularly which genetic and environmental contributors increase the risk of developing central sensitization in particular systems, exactly what triggers and sustains the phenomenon, and what is responsible in some individuals for its persistence. Nevertheless, the identification of the contribution of central sensitization to many unexplained clinical pain conditions has both provided a mechanistic explanation, and offered a therapeutic target. Conflict of interest There is no conflict of interest. Acknowledgements Supported by research funds from the NIH. I thank all my colleagues whose work has over the past 25 years contributed to the study of central sensitization but particularly Alban Latremoliere for his careful reading of the MS and Christian von Hehn for making the two figures. References [1] Aasvang EK, Brandsborg B, Jensen TS, Kehlet H. Heterogeneous sensory processing in persistent postherniotomy pain. Pain 2010;150: [2] Aggarwal VR, McBeth J, Zakrzewska JM, Lunt M, Macfarlane GJ. The epidemiology of chronic syndromes that are frequently unexplained: do they have common associated factors? Int J Epidemiol 2006;35: [3] Amital D, Herskovitz C, Fostick L, Silberman A, Doron Y, Zohar J, Itsekson A, Zolti M, Rubinow A, Amital H. The premenstrual syndrome and fibromyalgia similarities and common features. Clin Rev Allergy Immunol 2010;38: [4] Amr YM, Yousef AA. Evaluation of efficacy of the perioperative administration of Venlafaxine or gabapentin on acute and chronic postmastectomy pain. Clin J Pain 2010;26: [5] Amr YM, Yousef AA, Alzeftawy AE, Messbah WI, Saber AM. Effect of preincisional epidural fentanyl and bupivacaine on postthoracotomy pain and pulmonary function. Ann Thorac Surg 2010;89: [6] Andersen OK, Felsby S, Nicolaisen L, Bjerring P, Jensen TS, Arendt-Nielsen L. The effect of Ketamine on stimulation of primary and secondary hyperalgesic areas induced by capsaicin a double-blind, placebo-controlled, human experimental study. Pain 1996;66: [7] Arendt-Nielsen L, Nie H, Laursen MB, Laursen BS, Madeleine P, Simonsen OH, Graven-Nielsen T. Sensitization in patients with painful knee osteoarthritis. Pain 2010;149: [8] Arendt-Nielsen L, Petersen-Felix S, Fischer M, Bak P, Bjerring P, Zbinden AM. The effect of N-methyl-D-aspartate antagonist (ketamine) on single and repeated nociceptive stimuli: a placebo-controlled experimental human study. Anesth Analg 1995;81:63 8. [9] Arning K, Baron R. Evaluation of symptom heterogeneity in neuropathic pain using assessments of sensory functions. Neurotherapeutics 2009;6: [10] Arnold LD, Bachmann GA, Rosen R, Kelly S, Rhoads GG. Vulvodynia: characteristics and associations with comorbidities and quality of life. Obstet Gynecol 2006;107: [11] Attal N, Bouhassira D, Gautron M, Vaillant JN, Mitry E, Lepere C, Rougier P, Guirimand F. Thermal hyperalgesia as a marker of oxaliplatin neurotoxicity: a prospective quantified sensory assessment study. Pain 2009;144: [12] Baba H, Ji RR, Kohno T, Moore KA, Ataka T, Wakai A, Okamoto M, Woolf CJ. Removal of GABAergic inhibition facilitates polysynaptic A fiber-mediated excitatory transmission to the superficial spinal dorsal horn. Mol Cell Neurosci 2003;24: [13] Bajaj P, Graven-Nielsen T, Arendt-Nielsen L. Osteoarthritis and its association with muscle hyperalgesia: an experimental controlled study. Pain 2001;93: [14] Bamigboye AA, Hofmeyr GJ. Local anaesthetic wound infiltration and abdominal nerves block during caesarean section for postoperative pain relief. Cochrane Database Syst Rev 2009;3:CD [15] Bardin L, Gregoire S, Aliaga M, Malfetes N, Vitton O, Ladure P, Newman- Tancredi A, Depoortere R. Comparison of milnacipran, duloxetine and pregabalin in the formalin pain test and in a model of stress-induced ultrasonic vocalizations in rats. Neurosci Res 2010;66: [16] Baron R, Baron Y, Disbrow E, Roberts TP. Brain processing of capsaicininduced secondary hyperalgesia: a functional MRI study. Neurology 1999;53: [17] Baron R, Baron Y, Disbrow E, Roberts TP. Activation of the somatosensory cortex during Abeta-fiber mediated hyperalgesia. A MSI study. Brain Res 2000;871: [18] Basbaum AI, Fields HL. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu Rev Neurosci 1984;7: [19] Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst Rev 2006;1:CD [20] Bessou P, Perl ER. Response of cutaneous sensory units with unmyelinated fibers to noxious stimuli. J Neurophysiol 1969;32: [21] Beswick FB, Evanson JM. The heterosynaptic activation of motoneurones during post-tetanic potentiation. J Physiol 1955;128: [22] Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, Brenner GJ, Ji RR, Bean BP, Woolf CJ, Samad TA. Nociceptors are interleukin-1beta sensors. J Neurosci 2008;28: [23] Bishop T, Marchand F, Young AR, Lewin GR, McMahon SB. Ultraviolet-Binduced mechanical hyperalgesia: a role for peripheral sensitisation. Pain 2010;150: [24] Biurrun Manresa JA, Morch CD, Andersen OK. Long-term facilitation of nociceptive withdrawal reflexes following low-frequency conditioning electrical stimulation: a new model for central sensitization in humans. Eur J Pain 2010;14: [25] Bonica JJ. Anesthesiology in the People s Republic of China. Anesthesiology 1974;40: [26] Bradley LA, Kersh BC, DeBerry JJ, Deutsch G, Alarcon GA, McLain DA. Lessons from fibromyalgia: abnormal pain sensitivity in knee osteoarthritis. Novartis Found Symp 2004;260: [discussion ]. [27] Brock C, Andresen T, Frokjaer JB, Gale J, Olesen AE, Arendt-Nielsen L, Drewes AM. Central pain mechanisms following combined acid and capsaicin perfusion of the human oesophagus. Eur J Pain 2010;14: [28] Brown AG, Iggo A. A quantitative study of cutaneous receptors and afferent fibres in the cat and rabbit. J Physiol 1967;193:

17 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S11 [29] Brunelli M, Castellucci V, Kandel ER. Synaptic facilitation and behavioral sensitization in Aplysia: possible role of serotonin and cyclic AMP. Science 1976;194: [30] Buchgreitz L, Lyngberg AC, Bendtsen L, Jensen R. Frequency of headache is related to sensitization: a population study. Pain 2006;123: [31] Buchgreitz L, Lyngberg AC, Bendtsen L, Jensen R. Increased prevalence of tension-type headache over a 12-year period is related to increased pain sensitivity. A population study. Cephalalgia 2007;27: [32] Buchgreitz L, Lyngberg AC, Bendtsen L, Jensen R. Increased pain sensitivity is not a risk factor but a consequence of frequent headache: a population-based follow-up study. Pain 2008;137: [33] Burgess PR, Perl ER. Myelinated afferent fibres responding specifically to noxious stimulation of the skin. J Physiol 1967;190: [34] Burke SM, Shorten GD. Perioperative pregabalin improves pain and functional outcomes 3 months after lumbar discectomy. Anesth Analg 2010;110: [35] Burns D, Hill L, Essandoh M, Jarzembowski TM, Schuler HG, Janicki PK. Effect of valdecoxib pretreatment on pain and secondary hyperalgesia: a randomized controlled trial in healthy volunteers [ISRCTN , NCT ]. BMC Anesthesiol 2006;6:3. [36] Burstein R, Cutrer MF, Yarnitsky D. The development of cutaneous allodynia during a migraine attack clinical evidence for the sequential recruitment of spinal and supraspinal nociceptive neurons in migraine. Brain 2000;123: [37] Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, Bajwa ZH. An association between migraine and cutaneous allodynia. Ann Neurol 2000;47: [38] Buscher HC, van Goor H, Wilder-Smith OH. Effect of thoracoscopic splanchnic denervation on pain processing in chronic pancreatitis patients. Eur J Pain 2007;11: [39] Buscher HC, Wilder-Smith OH, van Goor H. Chronic pancreatitis patients show hyperalgesia of central origin: a pilot study. Eur J Pain 2006;10: [40] Cakit BD, Taskin S, Nacir B, Unlu I, Genc H, Erdem HR. Comorbidity of fibromyalgia and cervical myofascial pain syndrome. Clin Rheumatol 2010;29: [41] Campbell JN, Khan AA, Meyer RA, Raja SN. Responses to heat of C-fiber nociceptors in monkey are altered by injury in the receptive field but not by adjacent injury. Pain 1988;32: [42] Campbell JN, Raja SN, Meyer RA, Mackinnon SE. Myelinated afferents signal the hyperalgesia associated with nerve injury. Pain 1988;32: [43] Chacur M, Lambertz D, Hoheisel U, Mense S. Role of spinal microglia in myositis-induced central sensitisation: an immunohistochemical and behavioural study in rats. Eur J Pain 2009;13: [44] Chang YW, Waxman SG. Minocycline attenuates mechanical allodynia and central sensitization following peripheral second-degree burn injury. J Pain doi: /j.jpain [45] Chappell AS, Ossanna MJ, Liu-Seifert H, Iyengar S, Skljarevski V, Li LC, Bennett RM, Collins H. Duloxetine, a centrally acting analgesic, in the treatment of patients with osteoarthritis knee pain: a 13-week, randomized, placebocontrolled trial. Pain 2009;146: [46] Chen Z, Muscoli C, Doyle T, Bryant L, Cuzzocrea S, Mollace V, Mastroianni R, Masini E, Salvemini D. NMDA-receptor activation and nitroxidative regulation of the glutamatergic pathway during nociceptive processing. Pain 2010;149: [47] Chiang CY, Li Z, Dostrovsky JO, Sessle BJ. Central sensitization in medullary dorsal horn involves gap junctions and hemichannels. Neuroreport 2010;21: [48] Chiechio S, Zammataro M, Morales ME, Busceti CL, Drago F. Gereau RWt, Copani A, Nicoletti F. Epigenetic modulation of mglu2 receptors by histone deacetylase inhibitors in the treatment of inflammatory pain. Mol Pharmacol 2009;75: [49] Chizh BA, Gohring M, Troster A, Quartey GK, Schmelz M, Koppert W. Effects of oral pregabalin and aprepitant on pain and central sensitization in the electrical hyperalgesia model in human volunteers. Br J Anaesth 2007;98: [50] Cook AJ, Woolf CJ, Wall PD. Prolonged C-fibre mediated facilitation of the flexion reflex in the rat is not due to changes in afferent terminal or motoneurone excitability. Neurosci Lett 1986;70:91 6. [51] Cook AJ, Woolf CJ, Wall PD, McMahon SB. Dynamic receptive field plasticity in rat spinal cord dorsal horn following C-primary afferent input. Nature 1987;325: [52] Cooke L, Eliasziw M, Becker WJ. Cutaneous allodynia in transformed migraine patients. Headache 2007;47: [53] Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci 2009;32:1 32. [54] Coughlin SM, Karanicolas PJ, Emmerton-Coughlin HM, Kanbur B, Kanbur S, Colquhoun PH. Better late than never? Impact of local analgesia timing on postoperative pain in laparoscopic surgery: a systematic review and metaanalysis. Surg Endosc doi: /s [55] Cruccu G, Sommer C, Anand P, Attal N, Baron R, Garcia-Larrea L, Haanpaa M, Jensen TS, Serra J, Treede RD. EFNS guidelines on neuropathic pain assessment: revised Eur J Neurol 2010;17: [56] Cuadrado ML, Young WB, Fernandez-de-las-Penas C, Arias JA, Pareja JA. Migrainous corpalgia: body pain and allodynia associated with migraine attacks. Cephalalgia 2008;28: [57] Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Giani C, Zbinden AM, Radanov BP. Central hypersensitivity in chronic pain after whiplash injury. Clin J Pain 2001;17: [58] Dahl JB, Brennum J, Arendt-Nielsen L, Jensen TS, Kehlet H. The effect of preversus postinjury infiltration with lidocaine on thermal and mechanical hyperalgesia after heat injury to the skin. Pain 1993;53: [59] Dahl JB, Mathiesen O, Kehlet H. An expert opinion on postoperative pain management, with special reference to new developments. Expert Opin Pharmacother 2010;11: [60] Dahl JB, Moiniche S. Pre-emptive analgesia. Br Med Bull 2004;71: [61] de la Llave-Rincon AI, Fernandez-de-las-Penas C, Fernandez-Carnero J, Padua L, Arendt-Nielsen L, Pareja JA. Bilateral hand/wrist heat and cold hyperalgesia, but not hypoesthesia, in unilateral carpal tunnel syndrome. Exp Brain Res 2009;198: [62] de Tommaso M, Guido M, Libro G, Losito L, Sciruicchio V, Monetti C, Puca F. Abnormal brain processing of cutaneous pain in migraine patients during the attack. Neurosci Lett 2002;333: [63] de Tommaso M, Libro G, Guido M, Sciruicchio V, Losito L, Puca F. Heat pain thresholds and cerebral event-related potentials following painful CO2 laser stimulation in chronic tension-type headache. Pain 2003;104: [64] de Tommaso M, Sardaro M, Serpino C, Costantini F, Vecchio E, Prudenzano MP, Lamberti P, Livrea P. Fibromyalgia comorbidity in primary headaches. Cephalalgia 2009;29: [65] Desmeules JA, Cedraschi C, Rapiti E, Baumgartner E, Finckh A, Cohen P, Dayer P, Vischer TL. Neurophysiologic evidence for a central sensitization in patients with fibromyalgia. Arthritis Rheum 2003;48: [66] Diers M, Koeppe C, Yilmaz P, Thieme K, Markela-Lerenc J, Schiltenwolf M, van Ackern K, Flor H. Pain ratings and somatosensory evoked responses to repetitive intramuscular and intracutaneous stimulation in fibromyalgia syndrome. J Clin Neurophysiol 2008;25: [67] Dimcevski G, Staahl C, Andersen SD, Thorsgaard N, Funch-Jensen P, Arendt- Nielsen L, Drewes AM. Assessment of experimental pain from skin, muscle, and esophagus in patients with chronic pancreatitis. Pancreas 2007;35: [68] Dirks J, Moiniche S, Hilsted KL, Dahl JB. Mechanisms of postoperative pain: clinical indications for a contribution of central neuronal sensitization. Anesthesiology 2002;97: [69] Drewes AM, Pedersen J, Reddy H, Rasmussen K, Funch-Jensen P, Arendt- Nielsen L, Gregersen H. Central sensitization in patients with non-cardiac chest pain: a clinical experimental study. Scand J Gastroenterol 2006;41: [70] Duale C, Sibaud F, Guastella V, Vallet L, Gimbert YA, Taheri H, Filaire M, Schoeffler P, Dubray C. Perioperative ketamine does not prevent chronic pain after thoracotomy. Eur J Pain 2009;13: [71] Dullenkopf A, Muller R, Dillmann F, Wiedemeier P, Hegi TR, Gautschi S. An intraoperative pre-incision single dose of intravenous ketamine does not have an effect on postoperative analgesic requirements under clinical conditions. Anaesth Intensive Care 2009;37: [72] Dworkin RH, O Connor AB, Audette J, Baron R, Gourlay GK, Haanpaa ML, Kent JL, Krane EJ, Lebel AA, Levy RM, Mackey SC, Mayer J, Miaskowski C, Raja SN, Rice AS, Schmader KE, Stacey B, Stanos S, Treede RD, Turk DC, Walco GA, Wells CD. Recommendations for the pharmacological management of neuropathic pain: an overview and literature update. Mayo Clin Proc 2010;85:S3 S14. [73] Eichenberger U, Neff F, Sveticic G, Bjorgo S, Petersen-Felix S, Arendt-Nielsen L, Curatolo M. Chronic phantom limb pain: the effects of calcitonin, ketamine, and their combination on pain and sensory thresholds. Anesth Analg 2008;106: [table of contents]. [74] Estacion M, Harty TP, Choi JS, Tyrrell L, Dib-Hajj SD, Waxman SG. A sodium channel gene SCN9A polymorphism that increases nociceptor excitability. Ann Neurol 2009;66: [75] Fernandez-Carnero J, Fernandez-de-Las-Penas C, de la Llave-Rincon AI, Ge HY, Arendt-Nielsen L. Widespread mechanical pain hypersensitivity as sign of central sensitization in unilateral epicondylalgia: a blinded, controlled study. Clin J Pain 2009;25: [76] Fernandez-de-las-Penas C, de la Llave-Rincon AI, Fernandez-Carnero J, Cuadrado ML, Arendt-Nielsen L, Pareja JA. Bilateral widespread mechanical pain sensitivity in carpal tunnel syndrome: evidence of central processing in unilateral neuropathy. Brain 2009;132: [77] Fernandez-de-Las-Penas C, Galan-Del-Rio F, Alonso-Blanco C, Jimenez-Garcia R, Arendt-Nielsen L, Svensson P. Referred pain from muscle trigger points in the masticatory and neck-shoulder musculature in women with temporomandibular disorders. J Pain doi: /j.jpain [78] Fernandez-de-las-Penas C, Galan-del-Rio F, Fernandez-Carnero J, Pesquera J, Arendt-Nielsen L, Svensson P. Bilateral widespread mechanical pain sensitivity in women with myofascial temporomandibular disorder: evidence of impairment in central nociceptive processing. J Pain 2009;10: [79] Fernandez-de-Las-Penas C, Ge HY, Arendt-Nielsen L, Cuadrado ML, Pareja JA. The local and referred pain from myofascial trigger points in the temporalis muscle contributes to pain profile in chronic tension-type headache. Clin J Pain 2007;23: [80] Fernandez-de-las-Penas C, Ge HY, Cuadrado ML, Madeleine P, Pareja JA, Arendt-Nielsen L. Bilateral pressure pain sensitivity mapping of the temporalis muscle in chronic tension-type headache. Headache 2008;48:

18 S12 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 [81] Fernandez-de-Las-Penas C, Madeleine P, Caminero A, Cuadrado M, Arendt- Nielsen L, Pareja J. Generalized neck-shoulder hyperalgesia in chronic tension-type headache and unilateral migraine assessed by pressure pain sensitivity topographical maps of the trapezius muscle. Cephalalgia 2009;30: [82] Fernandez-de-Las-Penas C, Madeleine P, Martinez-Perez A, Arendt-Nielsen L, Jimenez-Garcia R, Pareja JA. Pressure pain sensitivity topograhical maps reveal bilateral hyperalgesia of the hands in patients with unilateral carpal tunnel syndrome. Arthritis Care Res (Hoboken) 2010;62: [83] Filatova E, Latysheva N, Kurenkov A. Evidence of persistent central sensitization in chronic headaches: a multi-method study. J Headache Pain 2008;9: [84] Foster DC, Dworkin RH, Wood RW. Effects of intradermal foot and forearm capsaicin injections in normal and vulvodynia-afflicted women. Pain 2005;117: [85] Freeman MD, Nystrom A, Centeno C. Chronic whiplash and central sensitization; an evaluation of the role of a myofascial trigger points in pain modulation. J Brachial Plex Peripher Nerve Inj 2009;4:2. [86] Freynhagen R, Rolke R, Baron R, Tolle TR, Rutjes AK, Schu S, Treede RD. Pseudoradicular and radicular low-back pain a disease continuum rather than different entities? Answers from quantitative sensory testing. Pain 2008;135: [87] Fuchs PN, Campbell JN, Meyer RA. Secondary hyperalgesia persists in capsaicin desensitized skin. Pain 2000;84: [88] Gao YJ, Zhang L, Samad OA, Suter MR, Yasuhiko K, Xu ZZ, Park JY, Lind AL, Ma Q, Ji RR. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci 2009;29: [89] Geisser ME, Strader Donnell C, Petzke F, Gracely RH, Clauw DJ, Williams DA. Comorbid somatic symptoms and functional status in patients with fibromyalgia and chronic fatigue syndrome: sensory amplification as a common mechanism. Psychosomatics 2008;49: [90] Gibson SJ, Littlejohn GO, Gorman MM, Helme RD, Granges G. Altered heat pain thresholds and cerebral event-related potentials following painful CO2 laser stimulation in subjects with fibromyalgia syndrome. Pain 1994;58: [91] Goncalves DA, Bigal ME, Jales LC, Camparis CM, Speciali JG. Headache and symptoms of temporomandibular disorder: an epidemiological study. Headache 2010;50: [92] Gottrup H, Juhl G, Kristensen AD, Lai R, Chizh BA, Brown J, Bach FW, Jensen TS. Chronic oral gabapentin reduces elements of central sensitization in human experimental hyperalgesia. Anesthesiology 2004;101: [93] Gottrup H, Kristensen AD, Bach FW, Jensen TS. Aftersensations in experimental and clinical hypersensitivity. Pain 2003;103: [94] Granot M, Buskila D, Granovsky Y, Sprecher E, Neumann L, Yarnitsky D. Simultaneous recording of late and ultra-late pain evoked potentials in fibromyalgia. Clin Neurophysiol 2001;112: [95] Graven-Nielsen T, Arendt-Nielsen L. Assessment of mechanisms in localized and widespread musculoskeletal pain. Nat Rev Rheumatol 2010;6: [96] Graven-Nielsen T, Aspegren Kendall S, Henriksson KG, Bengtsson M, Sorensen J, Johnson A, Gerdle B, Arendt-Nielsen L. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain 2000;85: [97] Griffin RS, Costigan M, Brenner GJ, Ma CH, Scholz J, Moss A, Allchorne AJ, Stahl GL, Woolf CJ. Complement induction in spinal cord microglia results in anaphylatoxin C5a-mediated pain hypersensitivity. J Neurosci 2007;27: [98] Guillemin R, Ling N, Burgus R, Bloom F, Segal D. Characterization of the endorphins, novel hypothalamic and neurohypophysial peptides with opiatelike activity: evidence that they induce profound behavioral changes. Psychoneuroendocrinology 1977;2: [99] Gwilym SE, Keltner JR, Warnaby CE, Carr AJ, Chizh B, Chessell I, Tracey I. Psychophysical and functional imaging evidence supporting the presence of central sensitization in a cohort of osteoarthritis patients. Arthritis Rheum 2009;61: [100] Han JS, Adwanikar H, Li Z, Ji G, Neugebauer V. Facilitation of synaptic transmission and pain responses by CGRP in the amygdala of normal rats. Mol Pain 2010;6:10. [101] Hardy JD, Wolff HG, Goodell H. Experimental evidence on the nature of cutaneous hyperalgesia. J Clin Invest 1950;29: [102] Hariharan S, Moseley H, Kumar A, Raju S. The effect of preemptive analgesia in postoperative pain relief a prospective double-blind randomized study. Pain Med 2009;10: [103] Harvey RJ, Depner UB, Wassle H, Ahmadi S, Heindl C, Reinold H, Smart TG, Harvey K, Schutz B, Abo-Salem OM, Zimmer A, Poisbeau P, Welzl H, Wolfer DP, Betz H, Zeilhofer HU, Muller U. GlyR alpha3: an essential target for spinal PGE2-mediated inflammatory pain sensitization. Science 2004;304: [104] Hathway GJ, Vega-Avelaira D, Moss A, Ingram R, Fitzgerald M. Brief, low frequency stimulation of rat peripheral C-fibres evokes prolonged microglialinduced central sensitization in adults but not in neonates. Pain 2009;144: [105] Hidalgo-Lozano A, Fernandez-de-las-Penas C, Alonso-Blanco C, Ge HY, Arendt-Nielsen L, Arroyo-Morales M. Muscle trigger points and pressure pain hyperalgesia in the shoulder muscles in patients with unilateral shoulder impingement: a blinded, controlled study. Exp Brain Res 2010;202: [106] Ho KY, Tay W, Yeo MC, Liu H, Yeo SJ, Chia SL, Lo NN. Duloxetine reduces morphine requirements after knee replacement surgery. Br J Anaesth 2010;105: [107] Hogeweg JA, Kuis W, Huygen AC, vos van Steenwijk C, Bernards AT, Oostendorp RA, Helders PJ. The pain threshold in juvenile chronic arthritis. Br J Rheumatol 1995;34:61 7. [108] Huge V, Lauchart M, Forderreuther S, Kaufhold W, Valet M, Azad SC, Beyer A, Magerl W. Interaction of hyperalgesia and sensory loss in complex regional pain syndrome type I (CRPS I). PLoS One 2008;3:e2742. [109] Hughes J, Smith TW, Kosterlitz HW, Fothergill LA, Morgan BA, Morris HR. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature 1975;258: [110] Iannetti GD, Zambreanu L, Wise RG, Buchanan TJ, Huggins JP, Smart TS, Vennart W, Tracey I. Pharmacological modulation of pain-related brain activity during normal and central sensitization states in humans. Proc Natl Acad Sci USA 2005;102: [111] Ifergane G, Buskila D, Simiseshvely N, Zeev K, Cohen H. Prevalence of fibromyalgia syndrome in migraine patients. Cephalalgia 2006;26: [112] Iggo A. Cutaneous mechanoreceptors with afferent C fibres. J Physiol 1960;152: [113] Ikeda H, Heinke B, Ruscheweyh R, Sandkuhler J. Synaptic plasticity in spinal lamina I projection neurons that mediate hyperalgesia. Science 2003;299: [114] Ikeda H, Stark J, Fischer H, Wagner M, Drdla R, Jager T, Sandkuhler J. Synaptic amplifier of inflammatory pain in the spinal dorsal horn. Science 2006;312: [115] Ilkjaer S, Dirks J, Brennum J, Wernberg M, Dahl JB. Effect of systemic N- methyl-d-aspartate receptor antagonist (dextromethorphan) on primary and secondary hyperalgesia in humans. Br J Anaesth 1997;79: [116] Ilkjaer S, Petersen KL, Brennum J, Wernberg M, Dahl JB. Effect of systemic N- methyl-d-aspartate receptor antagonist (ketamine) on primary and secondary hyperalgesia in humans. Br J Anaesth 1996;76: [117] Imamura M, Imamura ST, Kaziyama HH, Targino RA, Hsing WT, de Souza LP, Cutait MM, Fregni F, Camanho GL. Impact of nervous system hyperalgesia on pain, disability, and quality of life in patients with knee osteoarthritis: a controlled analysis. Arthritis Rheum 2008;59: [118] Iyengar S, Webster AA, Hemrick-Luecke SK, Xu JY, Simmons RM. Efficacy of duloxetine, a potent and balanced serotonin-norepinephrine reuptake inhibitor in persistent pain models in rats. J Pharmacol Exp Ther 2004;311: [119] Jensen MT, Petersen KL. Gender differences in pain and secondary hyperalgesia after heat/capsaicin sensitization in healthy volunteers. J Pain 2006;7: [120] Jensen R. Mechanisms of spontaneous tension-type headaches: an analysis of tenderness, pain thresholds and EMG. Pain 1996;64: [121] Ji RR, Baba H, Brenner GJ, Woolf CJ. Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci 1999;2: [122] Ji RR, Kohno T, Moore KA, Woolf CJ. Central sensitization and LTP: do pain and memory share similar mechanisms? Trends Neurosci 2003;26: [123] Ji RR, Samad TA, Jin SX, Schmoll R, Woolf CJ. P38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002;36: [124] Jones CK, Peters SC, Shannon HE. Efficacy of duloxetine, a potent and balanced serotonergic and noradrenergic reuptake inhibitor, in inflammatory and acute pain models in rodents. J Pharmacol Exp Ther 2005;312: [125] Jorum E, Warncke T, Stubhaug A. Cold allodynia and hyperalgesia in neuropathic pain: the effect of N-methyl-D-aspartate (NMDA) receptor antagonist ketamine a double-blind, cross-over comparison with alfentanil and placebo. Pain 2003;101: [126] Juhl GI, Jensen TS, Norholt SE, Svensson P. Central sensitization phenomena after third molar surgery: a quantitative sensory testing study. Eur J Pain 2008;12: [127] Kato K, Sullivan PF, Evengard B, Pedersen NL. Chronic widespread pain and its comorbidities: a population-based study. Arch Intern Med 2006;166: [128] Katz J, Cohen L, Schmid R, Chan VW, Wowk A. Postoperative morphine use and hyperalgesia are reduced by preoperative but not intraoperative epidural analgesia: implications for preemptive analgesia and the prevention of central sensitization. Anesthesiology 2003;98: [129] Kawasaki Y, Kohno T, Zhuang ZY, Brenner GJ, Wang H, Van Der Meer C, Befort K, Woolf CJ, Ji RR. Ionotropic and metabotropic receptors, protein kinase A, protein kinase C, and Src contribute to C-fiber-induced ERK activation and camp response element-binding protein phosphorylation in dorsal horn neurons, leading to central sensitization. J Neurosci 2004;24: [130] Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin- 6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 2008;28: [131] Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet 2006;367: [132] Khan AA, Owatz CB, Schindler WG, Schwartz SA, Keiser K, Hargreaves KM. Measurement of mechanical allodynia and local anesthetic efficacy in patients with irreversible pulpitis and acute periradicular periodontitis. J Endod 2007;33:

19 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S13 [133] Kim YH, Park CK, Back SK, Lee CJ, Hwang SJ, Bae YC, Na HS, Kim JS, Jung SJ, Oh SB. Membrane-delimited coupling of TRPV1 and mglur5 on presynaptic terminals of nociceptive neurons. J Neurosci 2009;29: [134] Kissin I. Preemptive analgesia: problems with assessment of clinical significance. Methods Mol Biol 2010;617: [135] Kitaj MB, Klink M. Pain thresholds in daily transformed migraine versus episodic migraine headache patients. Headache 2005;45: [136] Klein T, Stahn S, Magerl W, Treede RD. The role of heterosynaptic facilitation in long-term potentiation (LTP) of human pain sensation. Pain 2008;139: [137] Klumpp DJ, Rudick CN. Summation model of pelvic pain in interstitial cystitis. Nat Clin Pract Urol 2008;5: [138] Kocher L, Anton F, Reeh PW, Handwerker HO. The effect of carrageenaninduced inflammation on the sensitivity of unmyelinated skin nociceptors in the rat. Pain 1987;29: [139] Koltzenburg M, Lundberg LE, Torebjork HE. Dynamic and static components of mechanical hyperalgesia in human hairy skin. Pain 1992;51: [140] Koltzenburg M, Torebjork HE, Wahren LK. Nociceptor modulated central sensitization causes mechanical hyperalgesia in acute chemogenic and chronic neuropathic pain. Brain 1994;117: [141] Konttinen YT, Honkanen VE, Gronblad M, Keinonen M, Santavirta N, Santavirta S. The relation of extraarticular tenderness to inflammatory joint disease and personality in patients with rheumatoid arthritis. J Rheumatol 1992;19: [142] Koppert W, Dern SK, Sittl R, Albrecht S, Schuttler J, Schmelz M. A new model of electrically evoked pain and hyperalgesia in human skin: the effects of intravenous alfentanil, S(+)-ketamine, and lidocaine. Anesthesiology 2001;95: [143] Kupers R, Schneider FC, 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: [144] Kurland JE, Coyle WJ, Winkler A, Zable E. Prevalence of irritable bowel syndrome and depression in fibromyalgia. Dig Dis Sci 2006;51: [145] LaMotte RH, Shain CN, Simone DA, Tsai EF. Neurogenic hyperalgesia: psychophysical studies of underlying mechanisms. J Neurophysiol 1991;66: [146] LaMotte RH, Thalhammer JG, Torebjork HE, Robinson CJ. Peripheral neural mechanisms of cutaneous hyperalgesia following mild injury by heat. J Neurosci 1982;2: [147] Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain 2009;10: [148] Latremoliere A, Woolf CJ. Synaptic plasticity and central sensitization: author reply. J Pain 2010;11: [149] Lavand homme P, De Kock M. The use of intraoperative epidural or spinal analgesia modulates postoperative hyperalgesia and reduces residual pain after major abdominal surgery. Acta Anaesthesiol Belg 2006;57: [150] Le Bars D, Chitour D, Kraus E, Clot AM, Dickenson AH, Besson JM. The effect of systemic morphine upon diffuse noxious inhibitory controls (DNIC) in the rat: evidence for a lifting of certain descending inhibitory controls of dorsal horn convergent neurones. Brain Res 1981;215: [151] LeBlanc BW, Iwata M, Mallon AP, Rupasinghe CN, Goebel DJ, Marshall J, Spaller MR, Saab CY. A cyclic peptide targeted against PSD-95 blocks central sensitization and attenuates thermal hyperalgesia. Neuroscience 2010;167: [152] Lee KY, Chung K, Chung JM. Involvement of reactive oxygen species in longterm potentiation in the spinal cord dorsal horn. J Neurophysiol 2010;103: [153] Lee MC, Zambreanu L, Menon DK, Tracey I. Identifying brain activity specifically related to the maintenance and perceptual consequence of central sensitization in humans. J Neurosci 2008;28: [154] Li J, Baccei ML. Excitatory synapses in the rat superficial dorsal horn are strengthened following peripheral inflammation during early postnatal development. Pain 2009;143: [155] Lloyd DP. Post-tetanic potentiation of response in monosynaptic reflex pathways of the spinal cord. J Gen Physiol 1949;33: [156] Lorenz J, Grasedyck K, Bromm B. Middle and long latency somatosensory evoked potentials after painful laser stimulation in patients with fibromyalgia syndrome. Electroencephalogr Clin Neurophysiol 1996;100: [157] Lundblad H, Kreicbergs A, Jansson KA. Prediction of persistent pain after total knee replacement for osteoarthritis. J Bone Joint Surg Br 2008;90: [158] Magerl W, Wilk SH, Treede RD. Secondary hyperalgesia and perceptual windup following intradermal injection of capsaicin in humans. Pain 1998;74: [159] Maihofner C, Jesberger F, Seifert F, Kaltenhauser M. Cortical processing of mechanical hyperalgesia: a MEG study. Eur J Pain 2010;14: [160] Mannion RJ, Costigan M, Decosterd I, Amaya F, Ma QP, Holstege JC, Ji RR, Acheson A, Lindsay RM, Wilkinson GA, Woolf CJ. Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc Natl Acad Sci USA 1999;96: [161] Mathew NT, Kailasam J, Seifert T. Clinical recognition of allodynia in migraine. Neurology 2004;63: [162] Mathiesen O, Rasmussen ML, Dierking G, Lech K, Hilsted KL, Fomsgaard JS, Lose G, Dahl JB. Pregabalin and dexamethasone in combination with paracetamol for postoperative pain control after abdominal hysterectomy. A randomized clinical trial. Acta Anaesthesiol Scand 2009;53: [163] Melzack R, Wall PD. Pain mechanisms: a new theory. Science 1965;150: [164] Mendell LM, Wall PD. Responses of Single Dorsal Cord Cells to Peripheral Cutaneous Unmyelinated Fibres. Nature 1965;206:97 9. [165] Miraucourt LS, Moisset X, Dallel R, Voisin DL. Glycine inhibitory dysfunction induces a selectively dynamic, morphine-resistant, and neurokinin 1 receptor- independent mechanical allodynia. J Neurosci 2009;29: [166] Mohn C, Vassend O, Knardahl S. Experimental pain sensitivity in women with temporomandibular disorders and pain-free controls: the relationship to orofacial muscular contraction and cardiovascular responses. Clin J Pain 2008;24: [167] Moiniche S, Kehlet H, Dahl JB. A qualitative and quantitative systematic review of preemptive analgesia for postoperative pain relief: the role of timing of analgesia. Anesthesiology 2002;96: [168] Moore KA, Kohno T, Karchewski LA, Scholz J, Baba H, Woolf CJ. Partial peripheral nerve injury promotes a selective loss of GABAergic inhibition in the superficial dorsal horn of the spinal cord. J Neurosci 2002;22: [169] Munakata J, Naliboff B, Harraf F, Kodner A, Lembo T, Chang L, Silverman DH, Mayer EA. Repetitive sigmoid stimulation induces rectal hyperalgesia in patients with irritable bowel syndrome. Gastroenterology 1997;112: [170] Munro G. Pharmacological assessment of the rat formalin test utilizing the clinically used analgesic drugs gabapentin, lamotrigine, morphine, duloxetine, tramadol and ibuprofen: influence of low and high formalin concentrations. Eur J Pharmacol 2009;605: [171] Neumann S, Doubell TP, Leslie T, Woolf CJ. Inflammatory pain hypersensitivity mediated by phenotypic switch in myelinated primary sensory neurons. Nature 1996;384: [172] Norbury TA, MacGregor AJ, Urwin J, Spector TD, McMahon SB. Heritability of responses to painful stimuli in women: a classical twin study. Brain 2007;130: [173] O Neill S, Manniche C, Graven-Nielsen T, Arendt-Nielsen L. Generalized deeptissue hyperalgesia in patients with chronic low-back pain. Eur J Pain 2007;11: [174] Olesen SS, Brock C, Krarup AL, Funch-Jensen P, Arendt-Nielsen L, Wilder- Smith OH, Drewes AM. Descending Inhibitory Pain Modulation Is Impaired in Patients With Chronic Pancreatitis. Clin Gastroenterol Hepatol 2010;8: [175] Park JW, Clark GT, Kim YK, Chung JW. Analysis of thermal pain sensitivity and psychological profiles in different subgroups of TMD patients. Int J Oral Maxillofac Surg 2010;39: [176] Pedersen JL, Crawford ME, Dahl JB, Brennum J, Kehlet H. Effect of preemptive nerve block on inflammation and hyperalgesia after human thermal injury. Anesthesiology 1996;84: [177] Perl ER. Myelinated afferent fibres innervating the primate skin and their response to noxious stimuli. J Physiol 1968;197: [178] Perl ER, Kumazawa T, Lynn B, Kenins P. Sensitization of high threshold receptors with unmyelinated (C) afferent fibers. Prog Brain Res 1976;43: [179] Petzke F, Clauw DJ, Ambrose K, Khine A, Gracely RH. Increased pain sensitivity in fibromyalgia: effects of stimulus type and mode of presentation. Pain 2003;105: [180] Prescott SA, Sejnowski TJ, De Koninck Y. Reduction of anion reversal potential subverts the inhibitory control of firing rate in spinal lamina I neurons: towards a biophysical basis for neuropathic pain. Mol Pain 2006;2:32. [181] Rabben T. Effects of the NMDA receptor antagonist ketamine in electrically induced A delta-fiber pain. Methods Find Exp Clin Pharmacol 2000;22: [182] Randic M, Jiang MC, Cerne R. Long-term potentiation and long-term depression of primary afferent neurotransmission in the rat spinal cord. J Neurosci 1993;13: [183] Ranzolin A, Brenol JC, Bredemeier M, Guarienti J, Rizzatti M, Feldman D, Xavier RM. Association of concomitant fibromyalgia with worse disease activity score in 28 joints, health assessment questionnaire, and short form 36 scores in patients with rheumatoid arthritis. Arthritis Rheum 2009;61: [184] Remerand F, Le Tendre C, Baud A, Couvret C, Pourrat X, Favard L, Laffon M, Fusciardi J. The early and delayed analgesic effects of ketamine after total hip arthroplasty: a prospective, randomized, controlled, double-blind study. Anesth Analg 2009;109: [185] Riedl A, Schmidtmann M, Stengel A, Goebel M, Wisser AS, Klapp BF, Monnikes H. Somatic comorbidities of irritable bowel syndrome: a systematic analysis. J Psychosom Res 2008;64: [186] Rivera-Arconada I, Lopez-Garcia JA. Changes in membrane excitability and potassium currents in sensitized dorsal horn neurons of mice pups. J Neurosci 2010;30: [187] Rodriguez MA, Afari N, Buchwald DS. Evidence for overlap between urological and nonurological unexplained clinical conditions. J Urol 2009;182: [188] Rolke R, Baron R, Maier C, Tolle TR, Treede RD, Beyer A, Binder A, Birbaumer N, Birklein F, Botefur IC, Braune S, Flor H, Huge V, Klug R, Landwehrmeyer GB, Magerl W, Maihofner C, Rolko C, Schaub C, Scherens A, Sprenger T, Valet M, Wasserka B. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain 2006;123:

20 S14 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 [189] Samad TA, Moore KA, Sapirstein A, Billet S, Allchorne A, Poole S, Bonventre JV, Woolf CJ. Interleukin-1beta-mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature 2001;410: [190] Sandkuhler J. Understanding LTP in pain pathways. Mol Pain 2007;3:9. [191] Sandrini G, Antonaci F, Lanfranchi S, Milanov I, Danilov A, Nappi G. Asymmetrical reduction of the nociceptive flexion reflex threshold in cluster headache. Cephalalgia 2000;20: [192] Sang CN, Gracely RH, Max MB, Bennett GJ. Capsaicin-evoked mechanical allodynia and hyperalgesia cross nerve territories. Evidence for a central mechanism. Anesthesiology 1996;85: [193] Sarkar S, Aziz Q, Woolf CJ, Hobson AR, Thompson DG. Contribution of central sensitisation to the development of non-cardiac chest pain. Lancet 2000;356: [194] Sarkar S, Hobson AR, Furlong PL, Woolf CJ, Thompson DG, Aziz Q. Central neural mechanisms mediating human visceral hypersensitivity. Am J Physiol Gastrointest Liver Physiol 2001;281:G [195] Sarkar S, Thompson DG, Woolf CJ, Hobson AR, Millane T, Aziz Q. Patients with chest pain and occult gastroesophageal reflux demonstrate visceral pain hypersensitivity which may be partially responsive to acid suppression. Am J Gastroenterol 2004;99: [196] Sarkar S, Woolf CJ, Hobson AR, Thompson DG, Aziz Q. Perceptual wind-up in the human oesophagus is enhanced by central sensitisation. Gut 2006;55: [197] Scholz J, Mannion RJ, Hord DE, Griffin RS, Rawal B, Zheng H, Scoffings D, Phillips A, Guo J, Laing RJ, Abdi S, Decosterd I, Woolf CJ. A novel tool for the assessment of pain: validation in low back pain. PLoS Med 2009;6:e [198] Schulte H, Sollevi A, Segerdahl M. The distribution of hyperaemia induced by skin burn injury is not correlated with the development of secondary punctate hyperalgesia. J Pain 2004;5: [199] Schur EA, Afari N, Furberg H, Olarte M, Goldberg J, Sullivan PF, Buchwald D. Feeling bad in more ways than one: comorbidity patterns of medically unexplained and psychiatric conditions. J Gen Intern Med 2007;22: [200] Schwartzman RJ, Alexander GM, Grothusen JR, Paylor T, Reichenberger E, Perreault M. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain 2009;147: [201] Segerdahl M. Multiple dose gabapentin attenuates cutaneous pain and central sensitisation but not muscle pain in healthy volunteers. Pain 2006;125: [202] Sen H, Sizlan A, Yanarates O, Emirkadi H, Ozkan S, Dagli G, Turan A. A comparison of gabapentin and ketamine in acute and chronic pain after hysterectomy. Anesth Analg 2009;109: [203] Sesana L, Caprioli E, Marcazzan GM. Long period study of outdoor radon concentration in Milan and correlation between its temporal variations and dispersion properties of atmosphere. J Environ Radioact 2003;65: [204] Sethna NF, Liu M, Gracely R, Bennett GJ, Max MB. Analgesic and cognitive effects of intravenous ketamine-alfentanil combinations versus either drug alone after intradermal capsaicin in normal subjects. Anesth Analg 1998;86: [205] Seybold VS. The role of peptides in central sensitization. Handb Exp Pharmacol 2009;194: [206] Shenker NG, Haigh RC, Mapp PI, Harris N, Blake DR. Contralateral hyperalgesia and allodynia following intradermal capsaicin injection in man. Rheumatology (Oxford) 2008;47: [207] Shibata K, Yamane K, Iwata M. Change of excitability in brainstem and cortical visual processing in migraine exhibiting allodynia. Headache 2006;46: [208] Sivilotti L, Woolf CJ. The contribution of GABAA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J Neurophysiol 1994;72: [209] Sorensen J, Bengtsson A, Backman E, Henriksson KG, Bengtsson M. Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scand J Rheumatol 1995;24: [210] Sprenger T, Valet M, Woltmann R, Zimmer C, Freynhagen R, Kochs EF, Tolle TR, Wagner KJ. Imaging pain modulation by subanesthetic S-(+)-ketamine. Anesth Analg 2006;103: [211] Srbely JZ, Dickey JP, Bent LR, Lee D, Lowerison M. Capsaicin-induced central sensitization evokes segmental increases in trigger point sensitivity in humans. J Pain 2010;11: [212] Staahl C, Olesen AE, Andresen T, Arendt-Nielsen L, Drewes AM. Assessing efficacy of non-opioid analgesics in experimental pain models in healthy volunteers: an updated review. Br J Clin Pharmacol 2009;68: [213] Staud R. Are patients with systemic lupus erythematosus at increased risk for fibromyalgia? Curr Rheumatol Rep 2006;8: [214] Staud R, Bovee CE, Robinson ME, Price DD. Cutaneous C-fiber pain abnormalities of fibromyalgia patients are specifically related to temporal summation. Pain 2008;139: [215] Staud R, Cannon RC, Mauderli AP, Robinson ME, Price DD, Vierck Jr CJ. Temporal summation of pain from mechanical stimulation of muscle tissue in normal controls and subjects with fibromyalgia syndrome. Pain 2003;102: [216] Staud R, Craggs JG, Perlstein WM, Robinson ME, Price DD. Brain activity associated with slow temporal summation of C-fiber evoked pain in fibromyalgia patients and healthy controls. Eur J Pain 2008;12: [217] Staud R, Nagel S, Robinson ME, Price DD. Enhanced central pain processing of fibromyalgia patients is maintained by muscle afferent input: a randomized, double-blind, placebo-controlled study. Pain 2009;145: [218] Staud R, Price DD, Robinson ME, Mauderli AP, Vierck CJ. Maintenance of windup of second pain requires less frequent stimulation in fibromyalgia patients compared to normal controls. Pain 2004;110: [219] Staud R, Robinson ME, Price DD. Temporal summation of second pain and its maintenance are useful for characterizing widespread central sensitization of fibromyalgia patients. J Pain 2007;8: [220] Staud R, Robinson ME, Vierck Jr CJ, Cannon RC, Mauderli AP, Price DD. Ratings of experimental pain and pain-related negative affect predict clinical pain in patients with fibromyalgia syndrome. Pain 2003;105: [221] Staud R, Vierck CJ, Cannon RL, Mauderli AP, Price DD. Abnormal sensitization and temporal summation of second pain (wind-up) in patients with fibromyalgia syndrome. Pain 2001;91: [222] Sterling M, Jull G, Vicenzino B, Kenardy J. Characterization of acute whiplashassociated disorders. Spine (Phila Pa 1976) 2004;29: [223] Stubhaug A, Breivik H, Eide PK, Kreunen M, Foss A. Mapping of punctuate hyperalgesia around a surgical incision demonstrates that ketamine is a powerful suppressor of central sensitization to pain following surgery. Acta Anaesthesiol Scand 1997;41: [224] Sweet WH. Control of pain by direct electrical stimulation of peripheral nerves. Clin Neurosurg 1976;23: [225] Sycha T, Anzenhofer S, Lehr S, Schmetterer L, Chizh B, Eichler HG, Gustorff B. Rofecoxib attenuates both primary and secondary inflammatory hyperalgesia: a randomized, double blinded, placebo controlled crossover trial in the UV-B pain model. Pain 2005;113: [226] Takazawa T, MacDermott AB. Glycinergic and GABAergic tonic inhibition fine tune inhibitory control in regionally distinct subpopulations of dorsal horn neurons. J Physiol 2010;588: [227] Tao YX. Dorsal horn alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor trafficking in inflammatory pain. Anesthesiology 2010;112: [228] Tegeder I, Adolph J, Schmidt H, Woolf CJ, Geisslinger G, Lotsch J. Reduced hyperalgesia in homozygous carriers of a GTP cyclohydrolase 1 haplotype. Eur J Pain 2008;12: [229] Tegeder I, Costigan M, Griffin RS, Abele A, Belfer I, Schmidt H, Ehnert C, Nejim J, Marian C, Scholz J, Wu T, Allchorne A, Diatchenko L, Binshtok AM, Goldman D, Adolph J, Sama S, Atlas SJ, Carlezon WA, Parsegian A, Lotsch J, Fillingim RB, Maixner W, Geisslinger G, Max MB, Woolf CJ. GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence. Nat Med 2006;12: [230] Thompson SW, King AE, Woolf CJ. Activity-dependent changes in rat ventral horn neurons in vitro; summation of prolonged afferent evoked postsynaptic depolarizations produce a d-2-amino-5-phosphonovaleric acid sensitive windup. Eur J Neurosci 1990;2: [231] Thompson SW, Woolf CJ, Sivilotti LG. Small-caliber afferent inputs produce a heterosynaptic facilitation of the synaptic responses evoked by primary afferent A-fibers in the neonatal rat spinal cord in vitro. J Neurophysiol 1993;69: [232] Tietjen GE, Brandes JL, Peterlin BL, Eloff A, Dafer RM, Stein MR, Drexler E, Martin VT, Hutchinson S, Aurora SK, Recober A, Herial NA, Utley C, White L, Khuder SA. Allodynia in migraine: association with comorbid pain conditions. Headache 2009;49: [233] Torebjork HE, Lundberg LE, LaMotte RH. Central changes in processing of mechanoreceptive input in capsaicin-induced secondary hyperalgesia in humans. J Physiol 1992;448: [234] Torres Tortosa M, Roman Rico D. Risk of transmission of human immunodeficiency virus type 1 after accidents with needles from drug addicts, occurred in the community. Rev Clin Esp 1991;189:95 6. [235] Torresani C, Bellafiore S, De Panfilis G. Chronic urticaria is usually associated with fibromyalgia syndrome. Acta Derm Venereol 2009;89: [236] Trabulsi EJ, Patel J, Viscusi ER, Gomella LG, Lallas CD. Preemptive multimodal pain regimen reduces opioid analgesia for patients undergoing roboticassisted laparoscopic radical prostatectomy. Urology doi: / j.urology [237] Treede RD, Meyer RA, Raja SN, Campbell JN. Peripheral and central mechanisms of cutaneous hyperalgesia. Prog Neurobiol 1992;38: [238] Tuchman M, Barrett JA, Donevan S, Hedberg TG, Taylor CP. Central sensitization and Ca(V)alpha(2)delta ligands in chronic pain syndromes: pathologic processes and pharmacologic effect. J Pain doi: / j.jpain [239] Turini D, Beneforti P, Spinelli M, Malagutti S, Lazzeri M. Heat/burning sensation induced by topical application of capsaicin on perineal cutaneous area: new approach in diagnosis and treatment of chronic prostatitis/chronic pelvic pain syndrome? Urology 2006;67: [240] van den Broeke EN, van Rijn CM, Biurrun Manresa JA, Andersen OK, Arendt- Nielsen L, Wilder-Smith OH. Neurophysiological correlates of nociceptive heterosynaptic long-term potentiation in humans. J Neurophysiol 2010;103: [241] Vaneker M, Wilder-Smith OH, Schrombges P, de Man-Hermsen I, Oerlemans HM. Patients initially diagnosed as warm or cold CRPS 1 show differences in central sensory processing some eight years after diagnosis: a quantitative sensory testing study. Pain 2005;115:

21 C.J. Woolf / PAIN Ò 152 (2011) S2 S15 S15 [242] Vardeh D, Wang D, Costigan M, Lazarus M, Saper CB, Woolf CJ, Fitzgerald GA, Samad TA. COX2 in CNS neural cells mediates mechanical inflammatory pain hypersensitivity in mice. J Clin Invest 2009;119: [243] Vartiainen NV, Kirveskari E, Forss N. Central processing of tactile and nociceptive stimuli in complex regional pain syndrome. Clin Neurophysiol 2008;119: [244] Wall PD, Woolf CJ. Muscle but not cutaneous C-afferent input produces prolonged increases in the excitability of the flexion reflex in the rat. J Physiol 1984;356: [245] Wall PD, Woolf CJ. The brief and the prolonged facilitatory effects of unmyelinated afferent input on the rat spinal cord are independently influenced by peripheral nerve section. Neuroscience 1986;17: [246] Wang H, Bolognese J, Calder N, Baxendale J, Kehler A, Cummings C, Connell J, Herman G. Effect of morphine and pregabalin compared with diphenhydramine hydrochloride and placebo on hyperalgesia and allodynia induced by intradermal capsaicin in healthy male subjects. J Pain 2008;9: [247] Wang H, Kohno T, Amaya F, Brenner GJ, Ito N, Allchorne A, Ji RR, Woolf CJ. Bradykinin produces pain hypersensitivity by potentiating spinal cord glutamatergic synaptic transmission. J Neurosci 2005;25: [248] Wiesinger B, Malker H, Englund E, Wanman A. Back pain in relation to musculoskeletal disorders in the jaw-face: a matched case-control study. Pain 2007;131: [249] Wilder-Smith CH, Robert-Yap J. Abnormal endogenous pain modulation and somatic and visceral hypersensitivity in female patients with irritable bowel syndrome. World J Gastroenterol 2007;13: [250] Willert RP, Delaney C, Hobson AR, Thompson DG, Woolf CJ, Aziz Q. Constitutive cyclo-oxygenase-2 does not contribute to the development of human visceral pain hypersensitivity. Eur J Pain 2006;10: [251] Willert RP, Delaney C, Kelly K, Sharma A, Aziz Q, Hobson AR. Exploring the neurophysiological basis of chest wall allodynia induced by experimental oesophageal acidification - evidence of central sensitization. Neurogastroenterol Motil 2007;19: [252] Willert RP, Hobson AR, Delaney C, Hicks KJ, Dewit OE, Aziz Q. Neurokinin-1 receptor antagonism in a human model of visceral hypersensitivity. Aliment Pharmacol Ther 2007;25: [253] Willert RP, Woolf CJ, Hobson AR, Delaney C, Thompson DG, Aziz Q. The development and maintenance of human visceral pain hypersensitivity is dependent on the N-methyl-D-aspartate receptor. Gastroenterology 2004;126: [254] Witting N, Svensson P, Arendt-Nielsen L, Jensen TS. Repetitive intradermal capsaicin: differential effect on pain and areas of allodynia and punctate hyperalgesia. Somatosens Mot Res 2000;17:5 12. [255] Woolf CJ. Evidence for a central component of post-injury pain hypersensitivity. Nature 1983;306: [256] Woolf CJ. Long term alterations in the excitability of the flexion reflex produced by peripheral tissue injury in the chronic decerebrate rat. Pain 1984;18: [257] Woolf CJ. Generation of acute pain: central mechanisms. Br Med Bull 1991;47: [258] Woolf CJ. Pain: moving from symptom control toward mechanism-specific pharmacologic management. Ann Intern Med 2004;140: [259] Woolf CJ. Central sensitization: uncovering the relation between pain and plasticity. Anesthesiology 2007;106: [260] Woolf CJ, Chong MS. Preemptive analgesia treating postoperative pain by preventing the establishment of central sensitization. Anesth Analg 1993;77: [261] Woolf CJ, Costigan M. Transcriptional and posttranslational plasticity and the generation of inflammatory pain. Proc Natl Acad Sci USA 1999;96: [262] Woolf CJ, King AE. Physiology and morphology of multireceptive neurons with C-afferent fiber inputs in the deep dorsal horn of the rat lumbar spinal cord. J Neurophysiol 1987;58: [263] Woolf CJ, King AE. Subthreshold components of the cutaneous mechanoreceptive fields of dorsal horn neurons in the rat lumbar spinal cord. J Neurophysiol 1989;62: [264] Woolf CJ, King AE. Dynamic alterations in the cutaneous mechanoreceptive fields of dorsal horn neurons in the rat spinal cord. J Neurosci 1990;10: [265] Woolf CJ, Ma Q. Nociceptors noxious stimulus detectors. Neuron 2007;55: [266] Woolf CJ, Max MB. Mechanism-based pain diagnosis: issues for analgesic drug development. Anesthesiology 2001;95: [267] Woolf CJ, McMahon SB. Injury-induced plasticity of the flexor reflex in chronic decerebrate rats. Neuroscience 1985;16: [268] Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in pain. Science 2000;288: [269] Woolf CJ, Shortland P, Coggeshall RE. Peripheral nerve injury triggers central sprouting of myelinated afferents. Nature 1992;355:75 8. [270] Woolf CJ, Shortland P, Sivilotti LG. Sensitization of high mechanothreshold superficial dorsal horn and flexor motor neurones following chemosensitive primary afferent activation. Pain 1994;58: [271] Woolf CJ, Thompson SW. The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain 1991;44: [272] Woolf CJ, Thompson SW, King AE. Prolonged primary afferent induced alterations in dorsal horn neurones, an intracellular analysis in vivo and in vitro. J Physiol (Paris) 1988;83: [273] Woolf CJ, Wall PD. Relative effectiveness of C primary afferent fibers of different origins in evoking a prolonged facilitation of the flexor reflex in the rat. J Neurosci 1986;6: [274] Woolf CJ, Walters ET. Common patterns of plasticity contributing to nociceptive sensitization in mammals and Aplysia. Trends Neurosci 1991;14:74 8. [275] Xu YM, Ge HY, Arendt-Nielsen L. Sustained nociceptive mechanical stimulation of latent myofascial trigger point induces central sensitization in healthy subjects. J Pain doi: /j.jpain [276] Yang CC, Lee JC, Kromm BG, Ciol MA, Berger RE. Pain sensitization in male chronic pelvic pain syndrome: why are symptoms so difficult to treat? J Urol 2003;170:823 6 [discussion ]. [277] Zakine J, Samarcq D, Lorne E, Moubarak M, Montravers P, Beloucif S, Dupont H. Postoperative ketamine administration decreases morphine consumption in major abdominal surgery: a prospective, randomized, double-blind, controlled study. Anesth Analg 2008;106: [278] Zanette G, Cacciatori C, Tamburin S. Central sensitization in carpal tunnel syndrome with extraterritorial spread of sensory symptoms. Pain 2010;148: [279] Ziegler EA, Magerl W, Meyer RA, Treede RD. Secondary hyperalgesia to punctate mechanical stimuli. Central sensitization to A-fibre nociceptor input. Brain 1999;122:

22 PAIN Ò 152 (2011) S25 S32 Review Orofacial pain Kenneth M. Hargreaves Departments of Endodontics, Pharmacology, Physiology and Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article. 1. Introduction Pain in the oral and craniofacial system represents a major medical and social problem. Indeed, a U.S. Surgeon General s report on orofacial health concludes that,...oral health means much more than healthy teeth. It means being free of chronic oral-facial pain conditions... [172]. Community-based surveys indicate that many subjects commonly report pain in the orofacial region, with estimates of >39 million, or 22% of Americans older than 18 years of age, in the United States alone [108]. Other population-based surveys conducted in the United Kingdom [111,112], Germany [91], or regional pain care centers in the United States [54] report similar occurrence rates [135]. Importantly, chronic widespread body pain, patient sex and age, and psychosocial factors appear to serve as risk factors for chronic orofacial pain [1,2,92,99,138]. In addition to its high degree of prevalence, the reported intensities of various orofacial pain conditions are similar to that observed with many spinal pain disorders (Fig. 1). Moreover, orofacial pain is derived from many unique target tissues, such as the meninges, cornea, tooth pulp, oral/ nasal mucosa, and temporomandibular joint (Fig. 2), and thus has several unique physiologic characteristics compared with the spinal nociceptive system [23]. Given these considerations, it is not surprising that accurate diagnosis and effective management of orofacial pain conditions represents a significant health care problem. Publications in the field of orofacial pain demonstrate a steady increase over the last several decades (Fig. 3). This is a complex literature; a recent bibliometric analysis of orofacial pain articles published in indicated that 975 articles on orofacial pain were published in 275 journals from authors representing 54 countries [142]. Thus, orofacial pain disorders represent a complex constellation of conditions with an equally diverse literature base. Accordingly, this review will focus on a summary of major research foci on orofacial pain without attempting to provide a comprehensive review of the entire literature. 2. Physiologic studies on trigeminal pain Several reviews are available that document the historical development of physiologic research on the trigeminal nociceptive Address: Department of Endodontics, UTHSCSA, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA. Tel.: ; fax: address: Hargreaves@uthscsa.edu system [33,53,114,120,149]. More recent studies have characterized differences in electrophysiological [82], anatomical [10], or pharmacological [42,80] properties of trigeminal afferents innervating distinct target tissues. Collectively, many of these studies provide support for the hypothesis that target tissue interactions with trigeminal neuron terminals, via either soluble factors such as neurotrophins [48], or by integrin binding to extracellular matrix molecules [22,25], regulate the expression or trafficking of neuronal proteins, including ion channels and receptors [90,139] or second messenger signaling pathways [25]. Thus, the presence of unique target tissues innervated by trigeminal afferent fibers likely contributes to differences in the responsiveness of these neurons. A recent review characterizes differences between the trigeminal and spinal afferent systems under basal conditions [23]. Table 1 illustrates differences between the trigeminal and spinal systems after various forms of injury. Collectively, these studies indicate that the trigeminal system has many unique features that may contribute to distinct response patterns to tissue injury. The hypothesis of peripheral regulation of neuronal phenotype has been expanded by the recognition that estradiol selectively alters gene transcription in trigeminal neurons with increased expression of neuropeptides, such as prolactin, that are capable of sensitizing neuronal responses to capsaicin or noxious heat [49]. Additional studies have demonstrated that trigeminal peptidergic neurons undergo morphological changes ( sprouting ) in response to injury-induced inflammation in target tissues [33]. In contrast, there is a lack of sympathetic fiber sprouting in trigeminal ganglion cells, unlike the well-recognized occurrence in the spinal system [19,29,63]. Thus, an emerging body of evidence reveals the dynamic and specific responsiveness of the trigeminal system to either injury of its various target tissues or to the presence of certain gonadal steroids. Other studies have employed cultured trigeminal ganglia (TG) to evaluate cellular mechanisms of neuronal activation. For example, cannabinoids activate a calcineurin pathway leading to the rapid dephosphorylation and desensitization of transient receptor potential cation channel, subfamily V, member 1 (TRPV1), thereby contributing to an ionotropic mechanism for peripheral cannabinoid antinociception [4,5,89,133]. Moreover, accumulating evidence indicates a functional cross-desensitization between TRPV1 and TRP subfamily A, member 1 (TRPA1) on trigeminal neurons, possibly via formation of a heteromer [4,6,146,147]. Additional studies have used cultured TG to demonstrate that opioid receptors are /$36.00 Ó 2011 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved. doi: /j.pain

23 S26 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 Fig. 1. Comparison of pain intensity among spinal and orofacial pain disorders using the McGill Total Rank Pain Index (PRI[T]). The PRI(T) is an ordinal scale consisting of the sum of the ranks of words in each of the 20 sub-categories on the McGill Pain Questionnaire and ranges from 0 to 78. Data taken from: [14,35,71,73,116,117,150,164,176,183,184]. Cornea Nasal/oral Mucosa Dental Pulp Meninges TMJ TSNC expressed on sensory neurons but are not coupled to inhibitory signaling pathways under basal experimental conditions. Instead, pretreatment with arachidonic acid or with agonists to receptors coupled to Gaq signaling pathways (eg, bradykinin, trypsin) is required to induce the rapid development of a functional competence for opioid receptor signaling to Gai pathways leading to inhibition of neuronal activities [24,25,60,131,132]. These cellular findings are consistent with the observation that opioids have little efficacy for peripheral antinociception under basal conditions, but rapidly gain functional competence following injection of inflammatory mediators [145] or the development of inflammation. Recent studies have employed cultured trigeminal neurons to identify endogenous TRPV1 agonists [130,134]. Heating of isolated superfused peripheral tissue to a noxious temperature range leads to the release of oxidized linoleic acid metabolites (OLAMs), including 9- and 13-hydroxyoctadecadienoic acid (HODE). The administration of synthetic 9- and 13-HODE (and their oxoode metabolites) selectively activates TRPV1, leading to inward currents, increased accumulation of intracellular calcium, and triggering exocytosis of neuropeptides from TG neurons and thermal allodynia. These effects are blocked by TRPV1 antagonists and are Vg Vp Vo Vtr Vi Vc Fig. 2. Unique target tissues innervated by the trigeminal sensory system. Taken from Bereiter et al. [23], with permission. observed only in trigeminal neurons from wild-type mice but not TRPV1 knockouts [130]. Moreover, the intracellular delivery of compounds that block OLAM formation (eg, nordihydroguaiaretic acid) or a combination of anti-9- and anti-13-hode antibodies both significantly inhibit heat-evoked activation of trigeminal neurons. Collectively, these findings strongly implicate the OLAMs as a family of endogenous TRPV1 agonists. Interestingly, the pronounced effect of TRPV1 antagonists for blocking heat hyperalgesia in inflammation as well as mechanical allodynia (after intrathecal administration) has led to the hypothesis that an endogenous TRPV1 system might be activated under conditions of tissue injury. In support of this hypothesis, the administration of anti-olam antibodies produces a peripherally mediated thermal antinociception and a centrally mediated blockade of mechanical allodynia in the complete Freund s model of inflammation [130,134]. Thus, the OLAM system appears to contribute to acute heat detection by TRPV1 and to regulate more persistent conditions such as inflammatory pain. Future research directions may include preclinical studies focusing on mechanisms underlying differences between trigeminal and spinal pain conditions, mechanisms of sex-dependent differences in pain transduction and processing, and on the biological basis and pharmacological regulation of acute and chronic orofacial pain conditions. Many of these studies would be promoted by the development of standardized preclinical pain models and assessment methods. In addition to research on the biological mechanisms of nociceptive transmission, numerous clinical studies have described strong psychosocial/disability components to orofacial pain. Indeed, some diagnostic classification schemes differentiate the dimension of tissue contributions from psychosocial/disability factors [55] contributing to orofacial pain disorders. These studies demonstrate that the orofacial pain patient is confronted with a complex, multidimensional disorder that is best managed with appropriate treatment for all underlying factors [2,46,56,61,85,97,154]. 3. Studies on trigeminal inflammatory disorders Many translational studies have evaluated acute inflammatory injury to the trigeminal system. The dental impaction pain model has been developed as a standard clinical method for evaluating many analgesic drugs [43,45,115]. Other investigators have used this model of acute inflammatory pain to evaluate preemptive anesthesia [66,68], activation of endogenous opioid analgesic systems [70,78,104,105], local release of inflammatory mediators as collected by implanted microdialysis probes [50,67,77,162], other physiologic mechanisms [76], or the association of genetic polymorphisms with postoperative pain [95,96,98]. This clinical model has several notable advantages, including participation of relatively healthy subjects not taking concurrent drugs, standardized surgical procedures leading to reduced variance, and relatively large numbers of potential participants. Collectively, these studies on patients undergoing surgical dental extractions have contributed greatly to evaluation of analgesics, anesthetics, and anxiolytics, as well as basic biological research on human subjects. Other studies have focused on chronic inflammation of the oral and craniofacial region. Clinical studies on irreversible pulpitis in teeth ( toothache ) have demonstrated that this condition of bacterial-induced inflammation/necrosis is associated with significant changes in expression of ion channels [8,16,83,110,178,182], receptors [106], and neuropeptides [32,33]. Moreover, inflammation of a single tooth in patients appears sufficient to trigger central sensitization [59,94,127,151]. Animal studies on inflammation in the trigeminal region have demonstrated target-site-dependent differences in sensitization/activation [62,79,175] as well as sex-dependent differences in neuronal activities [20,126,168]. 21

24 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 S27 Fig. 3. Rates of articles published on orofacial pain. Data were acquired from a PubMed search (August 2010) using the search criteria of: (orofacial or trigeminal or temporomandibular or dental or tooth) and (pain or headache or hyperalgesia or allodynia or nociceptor or nociceptive). Future research directions on trigeminal inflammatory disorders may include preclinical and translational clinical studies focusing on mechanisms underlying the development and maintenance of inflammatory hyperalgesia/allodynia. Importantly, the clinical condition of pulpitis results in a very restricted pain locus (often within a tissue volume of <200 ul), intense pain reports [73], and dynamic neuronal and immunoplasticity. Thus, the pulpitis pain model is important not only from the perspective of high prevalence [108] and health care disparity [174], but also serves as a useful model for translational research [27]. 4. Studies on trigeminal neuropathic disorders The orofacial region has unique neuropathic pain disorders not seen in the spinal system, including trigeminal neuralgia and glossopharyngeal neuralgia [15]. Numerous clinical reports document these and other orofacial neuropathic or neuritic/neuralgic pain conditions and their responsiveness to surgical or pharmacological treatments [15,38,118,124,129,168,184]. Several etiologic factors appear to contribute to the development of neuropathic pain disorders. Proposed mechanisms include injury/compression to the trigeminal nerve, inflammatory insult (possibly including glial contributions), or infection with herpes virus [3,15,17,40,124, 125,177]. However, not all injuries to the trigeminal nerve lead to neuropathic pain disorders; indeed, the incidence of neuropathic pain after injury to orofacial structures is relatively low after dental treatment [34,109,137], facial trauma [17], orthognathic surgery [39], tooth extraction [26,36,143,173], or placement of dental implants [72]. This apparent resistance of the trigeminal system to development of neuropathic conditions is an interesting clinical observation that should prompt preclinical research comparing trigeminal to spinal afferent systems for susceptibility to neuropathic pain disorders. It is interesting that the trigeminal system appears programmed for periodic loss of innervated structures during postnatal development, with the shedding of 20 deciduous teeth per person, with minimal development of neuropathic pain conditions. Several risk factors for trigeminal neuralgia have been found, including multiple sclerosis [44,140] and hypertension [93]. Additional studies have reported changes in the expression of ion channels (eg, NaV1.3, 1.7, 1.8, TRPA1) in surgical biopsies collected from patients suffering from neuropathic orofacial pain [119,153]. Animal models of trigeminal neuropathic pain have been developed and include chronic constriction injury of the infraorbital nerve as well as transaction of the inferior alveolar nerve [3,177]. Interestingly, both preclinical and clinical studies have implicated constriction of peripheral nerves as an etiologic mechanism for inducing neuropathic pain via alteration in primary afferent functions [81], although certain cortical changes have been reported as well [28]. This has led to the development of clinical surgical decompression procedures to treat patients with trigeminal neuralgia [166,167]. Several preclinical studies have implicated ion channels and endothelin receptors as well as glial mechanisms in contributing to the development of these models of neuropathic pain conditions [11,40,81,125]. 5. Studies on chronic trigeminal myofascial and joint pain The diagnosis and management of many chronic orofacial pain conditions has been greatly hampered by confusion in determining etiologies from the temporomandibular joint versus myofascial sources. This has led to clinical studies difficult to interpret and diagnostic classifications that did not have a strong biological basis due to the lack of differentiation between joint and muscle contributions to the patient s pain condition. Clinical studies on myofascial pain or temporomandibular dysfunction (TMD) were considerably improved by the development of the Research Diagnostic Criteria [55,56], which highlighted the need for developing standardized diagnostic methods and definitions. Considerable evidence has been published demonstrating that patient sex/gender and exposure to sex steroids serve as risk factors for developing chronic orofacial pain conditions [61,65, ]. However, this is not observed in all studies, and other risk factors such as chronic widespread body pain, a prior history of physical abuse, or health anxiety have also been reported to be associated with the development of chronic orofacial pain disorders [1,55,61,92,100,107]. The reasons why some but not all studies detect sex/gender as a significant risk factor for orofacial pain disorders is not clear, but may be due to differences in patient populations, case definitions, or experimental approaches. Related preclinical studies have demonstrated that trigeminal neurons express estrogen receptors and undergo dramatic changes in gene expression [9,21,49] or firing rates [62] following exposure to estradiol. Other clinical studies have focused on synovial fluid levels of inflammatory mediators to test for other possible biological mechanisms [31,160] or have evaluated the role of peripheral glutamate receptors in triggering myofascial pain [7,12]. A very interesting approach is the application of genetics to patients with TMD. A haplotype of the catechol-o-methyltransferase gene in patients is associated with reduced responsiveness to experimental pain and to reduced risk for TMD [47]. Moreover, a mechanistic hypothesis for the protective effect of this haplotype has been advanced [121], and TMD patients with this catechol-o-methyltransferase haplotype respond with increased analgesia from drugs such as propranolol [169]. 6. Studies on other orofacial pain conditions Many other orofacial pain disorders also have been evaluated. The trigeminal autonomic cephalgias include cluster headache, 22

25 S28 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 Table 1 Comparison of the trigeminal and spinal afferent systems after injury. Marker Injury model Comparison Authors Galanin Axotomy TG DRG for upregulation Arvidsson et al. [13]; Zhang et al. [185] NPY Axotomy TG DRG for upregulation Arvidsson et al. [13]; Zhang et al. [185] Sympathetic fiber sprouting into ganglion and basket formation Axotomy or CCI TG: No DRG: Yes Bongenhielm et al. [29]; Benoliel et al. [19] Sympathetic fiber sprouting into NGF infusion icv X 14d DRG > TG Nauta et al. [122] ganglion SNS/PN3 = NaV1.8 Axotomy TG: Downregulation followed by Bongenhielm et al. [30] normalization DRG: Persistent downregulation Ankyrin(G) Axotomy TG: Persistent downregulation Bongenhielm et al. [30] Ectopic firing of afferents Axotomy TG < DRG Tal and Devor[165] Augmented excitability Axotomy TG DRG Tal and Devor [165]; Zhang et al. [186]; Cherkas et al. [37] Frequency and rhythmicity of Tight ligation of infraorbital vs sciatic DRG had significantly greater Tal and Devor [165] spontaneous discharges nerves spontaneous discharge rate than TG neurons for both myelinated and unmyelinated fibers. DRG afferents had rhythmic discharge rates (not seen with TG) Satellite glial cells Axotomy TG DRG for upregulation of GFPA, proliferation Woodham et al. [181]; Stephenson et al. [158]; Cherkas et al. [37] NOS Axotomy TG DRG for upregulation Hokfelt et al. [84] P2X3 & ATF-3 expression Partial axotomy TG DRG Tsuzuki et al. [171] GM3 ganglioside. Knockout GM2/GD2 and the GD3 Facial wounding > Rest of the body Inoue et al. [87] synthase gene With peripheral nerve degeneration Peripheral chromatolysis LiCl TG DRG Levine et al. (2004) Sensory neuropathy with neuronal degeneration Sjogren s Syndrome TG DRG Malinow et al. [113] Infectivity of contralateral ganglia HSV polypeptide ICP4 (VP175) expression in ganglia Viral replication and degradation of host cells, mrna Herpes simplex virus-1 (HSV) infection Herpes simplex virus-1 (HSV) infection Herpes simplex virus-1 (HSV) infection 70% of TG contralateral to side of HSV Thackray et al. [170] injection produced infections after inoculation, whereas only 10% of contralateral DRG produced infections. TG DRG Pepose et al. [136] TG DRG with wild-type HSV more virulent in both ganglia than HSV mutants lacking virion host shutoff (vhs) protein Smith et al. [156] Infectivity of ganglia Simian varicella virus (SVV) TG DRG White et al. [179] Substance P in ganglia Streptozotocin-diabetes TG had 26% reduction (P < 0.01), but Robinson et al. [144] DRG = 11% non-significant reduction Substance P in ganglia mf rat (mutilated foot; an autosomal DRG < TG Scaravilli [148] recessive sensory neuropathy with reduced pain responsiveness Caspase-3 mediated neuronal Knockout of Rb (retinoblastoma TG DRG for protection from Simpson et al. [152] apoptosis tumor suppressor protein) apoptosis in double knockout of Rb and caspase-3 compared to single Rb knockout Number of neurons in ganglia TRKa knockout TG DRG for extensive neuronal loss Smeyne et al. [155] Reactivation of virus HSV mutant with gamma 34.5 gene TG > resistant to reactivation than Spivack et al. [157] deletion DRG Wide-spread numbness and pain 4 12 d after antibiotic treatment Acute sensory neuronopathy syndrome in humans TG DRG Sterman et al. [159] Pain Trigeminal neuralgia in humans TG: Yes (max/mand divisions > ophthalmic) DRG: No equivalent Spontaneous behavior Formalin OVX females exhibited significantly greater increase in formalin hyperalgesia after orofacial injection (upper lip) compared to hind paw injection. Result is consistent with hypothesis of a difference in sex steroid regulation of nociception between TG and DRG systems Jannetta [88]; Sweet [161]; Wilkins [180]; Goya et al. [69]; Hamlyn [74,75]; Tacconi and Miles [163] Pajot et al. [128] SNS, sensory neuron specific; PN3, peripheral nerve sodium channel type 3; NOS, nitric oxide synthase; P2X3, purinoceptor 3; ICP4, infected cell protein 4; TG, trigeminal ganglia; DRG, dorsal root ganglia; NPY, neuropeptide Y; CCI, chronic constriction injury; NGF, nerve growth factor; NOA, ATF-3, activating transcription factor 3; OVX, ovariectomized. paroxysmal hemicrania, and unilateral neuralgiform headaches [41]. This collection of pain disorders is characterized by unilateral head pain in association with autonomic features such as tearing and conjunctival involvement, and considerable research has shed light on pain referral patterns and issues related to proper diagnosis and treatment [18,51,58,64,86]. Most cases of trigeminal auto- 23

26 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 S29 nomic cephalgia reflect primary headaches, although rare cases may be associated with pituitary tumors [41]. Pain is a major aspect of oral cancer [57] and often represents the initial symptom that prompts patients to seek health providers. Pain due to oral cancer may be due to soluble factors released from tumor cells, a localized inflammatory response to the tumor, or even nerve entrapment. Several recent studies have implicated the endothelin system and proteases (eg, PAR-2 receptor activation) in mediating mechanical allodynia experimental models of oral cancer pain [52,141]. Burning mouth syndrome is a rare disorder, commonly characterized by spontaneous burning pain and mechanical allodynia. Although idiopathic, it has many features of neuropathic pain and has been reported to be associated with altered peripheral expression of voltage-gated sodium channels [16]. 7. Discussion Orofacial pain disorders comprise a major and expensive component of health care and collectively have a high prevalence rate, a large range in pain intensity with a commensurate, often devastating impact on quality of life [149]. Although there are many common aspects of pain transduction and processing between the trigeminal and spinal systems, there are numerous examples of unique features in the peripheral and central components of the trigeminal pain system. Accordingly, ongoing basic and clinical research focused on acute and chronic orofacial pain conditions is required to understand the unique features of this pain system and to develop and evaluate better ways to treat patients with orofacial pain. A major barrier for improved patient care and translational research is the lack of validated diagnostic criteria. Although efforts have been made to classify TMD patients with the Research Diagnostic Criteria for TMD, headache patients with the International Headache Society criteria, and orofacial pain with the American Academy of Orofacial Pain standards, clinical research indicates that each of these 3 methods is incomplete for comprehensive diagnosis of orofacial pain patients [18]. Thus, further research is critically required to establish a comprehensive, sensitive, and specific diagnostic classification scheme for all orofacial pain patients. This would provide a critical contribution to practitioners and foster the development of a powerful dataset for clinical research. In addition, recent studies have incorporated quality-of-life indices, which provide important additional information on clinical outcomes [123]. Taken together, orofacial pain conditions represent a highly prevalent spectrum of pain disorders with pain intensities similar to those observed with many chronic spinal pain conditions. However, the unique anatomical, biochemical, and associated psychosocial components provide compelling evidence for specific research focused on orofacial pain disorders. Conflict of interest statement The author denies any conflicts of interest. Acknowledgement This work was supported in part by R01 NS72890, R01 NS58655, R01 DA19585, and NCRR U54RR References [1] Aggarwal VR, Macfarlane GJ, Farragher TM, McBeth J. Risk factors for onset of chronic oro-facial pain results of the North Cheshire oro-facial pain prospective population study. Pain 2010;149: [2] Aggarwal VR, Macfarlane TV, Macfarlane GJ. Why is pain more common amongst people living in areas of low socio-economic status? A populationbased cross-sectional study. Br Dent J 2003;194:383 7 [discussion 380]. [3] Ahn DK, Lim EJ, Kim BC, Yang GY, Lee MK, Ju JS, Han SR, Bae YC. Compression of the trigeminal ganglion produces prolonged nociceptive behavior in rats. Eur J Pain 2009;13: [4] Akopian AN, Ruparel NB, Jeske NA, Hargreaves KM. Transient receptor potential TRPA1 channel desensitization in sensory neurons is agonist dependent and regulated by TRPV1-directed internalization. J Physiol 2007;583: [5] Akopian AN, Ruparel NB, Jeske NA, Patwardhan A, Hargreaves KM. Role of ionotropic cannabinoid receptors in peripheral antinociception and antihyperalgesia. Trends Pharmacol Sci 2009;30: [6] Akopian AN, Ruparel NB, Patwardhan A, Hargreaves KM. Cannabinoids desensitize capsaicin and mustard oil responses in sensory neurons via TRPA1 activation. J Neurosci 2008;28: [7] Alstergren P, Ernberg M, Nilsson M, Hajati AK, Sessle BJ, Kopp S. Glutamate-induced temporomandibular joint pain in healthy individuals is partially mediated by peripheral NMDA receptors. J Orofac Pain 2010;24: [8] Alvarado LT, Perry GM, Hargreaves KM, Henry MA. TRPM8 Axonal expression is decreased in painful human teeth with irreversible pulpitis and cold hyperalgesia. J Endod 2007;33: [9] Amandusson A, Blomqvist A. Estrogen receptor-alpha expression in nociceptive-responsive neurons in the medullary dorsal horn of the female rat. Eur J Pain 2010;14: [10] Ambalavanar R, Moritani M, Dessem D. Trigeminal P2X3 receptor expression differs from dorsal root ganglion and is modulated by deep tissue inflammation. Pain 2005;117: [11] Amir R, Argoff CE, Bennett GJ, Cummins TR, Durieux ME, Gerner P, Gold MS, Porreca F, Strichartz GR. The role of sodium channels in chronic inflammatory and neuropathic pain. J Pain 2006;7:S1 S29. [12] Arendt-Nielsen L, Svensson P, Sessle BJ, Cairns BE, Wang K. Interactions between glutamate and capsaicin in inducing muscle pain and sensitization in humans. Eur J Pain 2008;12: [13] Arvidsson J, Raappana P, Diez M, Hokfelt T. Expression of neuropeptides in the rat mesencephalic trigeminal nucleus after peripheral axotomy. Neuroreport 1994;5: [14] Barlas P, Craig JA, Robinson J, Walsh DM, Baxter GD, Allen JM. Managing delayed-onset muscle soreness: lack of effect of selected oral systemic analgesics. Arch Phys Med Rehabil 2000;81: [15] Baron R, Binder A, Wasner G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol 2010;9: [16] Beneng K, Renton T, Yilmaz Z, Yiangou Y, Anand P. Sodium channel Na v 1.7 immunoreactivity in painful human dental pulp and burning mouth syndrome. BMC Neurosci 2010;11:71. [17] Benoliel R, Birenboim R, Regev E, Eliav E. Neurosensory changes in the infraorbital nerve following zygomatic fractures. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2005;99: [18] Benoliel R, Birman N, Eliav E, Sharav Y. The International Classification of Headache Disorders: accurate diagnosis of orofacial pain? Cephalalgia 2008;28: [19] Benoliel R, Eliav E, Tal M. No sympathetic nerve sprouting in rat trigeminal ganglion following painful and non-painful infraorbital nerve neuropathy. Neurosci Lett 2001;297: [20] Bereiter DA, Benetti AP. Amino acid release at the spinomedullary junction after inflammation of the TMJ region in male and female rats. Pain 2006;126: [21] Bereiter DA, Cioffi JL, Bereiter DF. Oestrogen receptor-immunoreactive neurons in the trigeminal sensory system of male and cycling female rats. Arch Oral Biol 2005;50: [22] Bereiter DA, Cioffi JL, Bereiter DF, Zardeneta G, Milam SB. Local blockade of integrins in the temporomandibular joint region reduces Fos-positive neurons in trigeminal subnucleus caudalis of female rats produced by jaw movement. Pain 2006;125: [23] Bereiter DA, Hargreaves KM, Hu JW. Trigeminal mechanisms of nociception: peripheral and brainstem organization. In: Basbaum AI, Kaneko A, Shepherd GM, Westheimer G, Albright T, Masland RH, Dallos P, Oertel D, Firestein SJ, Beauchamp GK, Bushnell MC, Kaas JH, Gardner EP, editors. The senses: a comprehensive reference. San Diego: Academic Press; p [24] Berg KA, Patwardhan AM, Sanchez TA, Silva YM, Hargreaves KM, Clarke WP. Rapid modulation of micro-opioid receptor signaling in primary sensory neurons. J Pharmacol Exp Ther 2007;321: [25] Berg KA, Zardeneta G, Hargreaves KM, Clarke WP, Milam SB. Integrins regulate opioid receptor signaling in trigeminal ganglion neurons. Neuroscience 2007;144: [26] Berge TI. Incidence of chronic neuropathic pain subsequent to surgical removal of impacted third molars. Acta Odontol Scand 2002;60: [27] Billinton A. Human dental pulp as a source of native functional nociceptor pharmacology. Pain 2009;144:227. [28] Blatow M, Nennig E, Sarpaczki E, Reinhardt J, Schlieter M, Herweh C, Rasche D, Tronnier VM, Sartor K, Stippich C. Altered somatosensory processing in trigeminal neuralgia. Hum Brain Mapp 2009;30: [29] Bongenhielm U, Boissonade FM, Westermark A, Robinson PP, Fried K. Sympathetic nerve sprouting fails to occur in the trigeminal ganglion after peripheral nerve injury in the rat. Pain 1999;82:

27 S30 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 [30] Bongenhielm U, Nosrat CA, Nosrat I, Eriksson J, Fjell J, Fried K. Expression of sodium channel SNS/PN3 and ankyrin(g) mrnas in the trigeminal ganglion after inferior alveolar nerve injury in the rat. Exp Neurol 2000;164: [31] Bouloux GF. Temporomandibular joint pain and synovial fluid analysis: a review of the literature. J Oral Maxillofac Surg 2009;67: [32] Bowles WR, Withrow JC, Lepinski AM, Hargreaves KM. Tissue levels of immunoreactive substance P are increased in patients with irreversible pulpitis. J Endod 2003;29: [33] Byers MR, Narhi MV. Dental injury models: experimental tools for understanding neuroinflammatory interactions and polymodal nociceptor functions. Crit Rev Oral Biol Med 1999;10:4 39. [34] Campbell RL, Parks KW, Dodds RN. Chronic facial pain associated with endodontic therapy. Oral Surg Oral Med Oral Pathol 1990;69: [35] Campbell WI, Kendrick RW. Pre-emptive analgesia using local anaesthesia: a study in bilaterally symmetrical surgery. Br J Anaesth 1997;79: [36] Carmichael FA, McGowan DA. Incidence of nerve damage following third molar removal: a West of Scotland Oral Surgery Research Group study. Br J Oral Maxillofac Surg 1992;30: [37] Cherkas PS, Huang TY, Pannicke T, Tal M, Reichenbach A, Hanani M. The effects of axotomy on neurons and satellite glial cells in mouse trigeminal ganglion. Pain 2004;110: [38] Cheshire Jr WP. Defining the role for gabapentin in the treatment of trigeminal neuralgia: a retrospective study. J Pain 2002;3: [39] Cheung LK, Lo J. The long-term clinical morbidity of mandibular step osteotomy. Int J Adult Orthodon Orthognath Surg 2002;17: [40] Chichorro JG, Zampronio AR, Cabrini DA, Franco CRC, Rae GA. Mechanisms operated by endothelin ETA and ETB receptors in the trigeminal ganglion contribute to orofacial thermal hyperalgesia induced by infraorbital nerve constriction in rats. Neuropeptides 2009;43: [41] Cittadini E, Matharu MS. Symptomatic trigeminal autonomic cephalalgias [Review]. Neurologist 2009;15: [42] Cook SP, Vulchanova L, Hargreaves KM, Elde R, McCleskey EW. Distinct ATP receptors on pain-sensing and stretch-sensing neurons. Nature 1997;387: [43] Cooper SA, Desjardins PJ. The value of the dental impaction pain model in drug development. Methods Mol Biol 2010;617: [44] Cruccu G, Biasiotta A, Di Rezze S, Fiorelli M, Galeotti F, Innocenti P, Mameli S, Millefiorini E, Truini A. Trigeminal neuralgia and pain related to multiple sclerosis. Pain 2009;143: [45] Derry C, Derry S, Moore RA, McQuay HJ. Single dose oral ibuprofen for acute postoperative pain in adults. Cochrane Database Syst Rev 2009:CD [46] Diatchenko L, Anderson AD, Slade GD, Fillingim RB, Shabalina SA, Higgins TJ, Sama S, Belfer I, Goldman D, Max MB, Weir BS, Maixner W. Three major haplotypes of the beta2 adrenergic receptor define psychological profile, blood pressure, and the risk for development of a common musculoskeletal pain disorder. Am J Med Genet B Neuropsychiatr Genet 2006;141B: [47] Diatchenko L, Slade GD, Nackley AG, Bhalang K, Sigurdsson A, Belfer I, Goldman D, Xu K, Shabalina SA, Shagin D, Max MB, Makarov SS, Maixner W. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum Mol Genet 2005;14: [48] Diogenes A, Akopian AN, Hargreaves KM. NGF up-regulates TRPA1: implications for orofacial pain. J Dent Res 2007;86: [49] Diogenes A, Patwardhan AM, Jeske NA, Ruparel NB, Goffin V, Akopian AN, Hargreaves KM. Prolactin modulates TRPV1 in female rat trigeminal sensory neurons. J Neurosci 2006;26: [50] Dionne RA, Gordon SM, Rowan J, Kent A, Brahim JS. Dexamethasone suppresses peripheral prostanoid levels without analgesia in a clinical model of acute inflammation. J Oral Maxillofac Surg 2003;61: [51] Dodick DW. Migraine with isolated facial pain: a diagnostic challenge. Cephalalgia 2007;27: [52] Dolan JC, Lam DK, Achdjian SH, Schmidt BL. The dolognawmeter: a novel instrument and assay to quantify nociception in rodent models of orofacial pain. J Neurosci Methods 2010;187: [53] Dubner R, Bennett GJ. Spinal and trigeminal mechanisms of nociception. Annu Rev Neurosci 1983;6: [54] Dworkin SF, Huggins KH, LeResche L, Von Korff M, Howard J, Truelove E, Sommers E. Epidemiology of signs and symptoms in temporomandibular disorders: clinical signs in cases and controls. J Am Dent Assoc 1990;120: [55] Dworkin SF, LeResche L. Research diagnostic criteria for temporomandibular disorders: review, criteria, examinations and specifications, critique. J Craniomandib Disord 1992;6: [56] Dworkin SF, Sherman J, Mancl L, Ohrbach R, LeResche L, Truelove E. Reliability, validity, and clinical utility of the research diagnostic criteria for Temporomandibular Disorders Axis II Scales: depression, non-specific physical symptoms, and graded chronic pain. J Orofac Pain 2002;16: [57] Epstein JB, Hong C, Logan RM, Barasch A, Gordon SM, Oberlee-Edwards L, McGuire D, Napenas JJ, Elting LS, Spijkervet FKL, Brennan MT. A systematic review of orofacial pain in patients receiving cancer therapy. Support Care Cancer 2010;18: [58] Eross E, Dodick D, Eross M. The Sinus, Allergy and Migraine Study (SAMS). Headache 2007;47: [59] Falace DA, Reid K, Rayens MK. The influence of deep (odontogenic) pain intensity, quality, and duration on the incidence and characteristics of referred orofacial pain. J Orofac Pain 1996;10: [60] Fehrenbacher JC, Sun XX, Locke EE, Henry MA, Hargreaves KM. Capsaicinevoked icgrp release from human dental pulp: a model system for the study of peripheral neuropeptide secretion in normal healthy tissue. Pain 2009;144: [61] Fillingim RB, Maixner W, Sigurdsson A, Kincaid S. Sexual and physical abuse history in subjects with temporomandibular disorders: relationship to clinical variables, pain sensitivity, and psychologic factors. J Orofac Pain 1997;11: [62] Flake NM, Bonebreak DB, Gold MS. Estrogen and inflammation increase the excitability of rat temporomandibular joint afferent neurons. J Neurophysiol 2005;93: [63] Fried K, Bongenhielm U, Boissonade FM, Robinson PP. Nerve injury-induced pain in the trigeminal system. Neuroscientist 2001;7: [64] Gaul C, Sandor PS, Galli U, Palla S, Ettlin DA. Orofacial migraine. Cephalalgia 2007;27: [65] Goncalves DA, Dal Fabbro AL, Campos JA, Bigal ME, Speciali JG. Symptoms of temporomandibular disorders in the population: an epidemiological study. J Orofac Pain 2010;24: [66] Gordon SM, Brahim JS, Dubner R, McCullagh LM, Sang C, Dionne RA. Attenuation of pain in a randomized trial by suppression of peripheral nociceptive activity in the immediate postoperative period. Anesth Analg 2010;95: [67] Gordon SM, Brahim JS, Rowan J, Kent A, Dionne RA. Peripheral prostanoid levels and nonsteroidal anti-inflammatory drug analgesia: replicate clinical trials in a tissue injury model. Clin Pharmacol Ther 2002;72: [68] Gordon SM, Dionne RA, Brahim J, Jabir F, Dubner R. Blockade of peripheral neuronal barrage reduces postoperative pain. Pain 1997;70: [69] Goya T, Wakisaka S, Kinoshita K. Microvascular decompression for trigeminal neuralgia with special reference to delayed recurrence. Neurol Med Chi (Tokyo) 1990;30: [70] Gracely RH, Dubner R, Wolskee PJ, Deeter WR. Placebo and naloxone can alter post-surgical pain by separate mechanisms. Nature 1983;306: [71] Graham C, Bond SS, Gerkovich MM, Cook MR. Use of the McGill pain questionnaire in the assessment of cancer pain: replicability and consistency. Pain 1980;8: [72] Gregg JM. Neuropathic complications of mandibular implant surgery: review and case presentations. Ann R Australas Coll Dent Surg 2000;15: [73] Grushka M, Sessle BJ. Applicability of the McGill Pain Questionnaire to the differentiation of toothache pain. Pain 1984;19: [74] Hamlyn PJ. Neurovascular relationships in the posterior cranial fossa, with special reference to trigeminal neuralgia. 1. Review of the literature and development of a new method of vascular injection-filling in cadaveric controls. Clin Anat 1997;10: [75] Hamlyn PJ. Neurovascular relationships in the posterior cranial fossa, with special reference to trigeminal neuralgia. 2. Neurovascular compression of the trigeminal nerve in cadaveric controls and patients with trigeminal neuralgia: quantification and influence of method. Clin Anat 1997;10: [76] Hamza M, Wang X-M, Adam A, Brahim JS, Rowan JS, Carmona GN, Dionne RA. Kinin B1 receptors contributes to acute pain following minor surgery in humans. Mol Pain 2010;6:12. [77] Hargreaves KM, Costello A. Glucocorticoids suppress levels of immunoreactive bradykinin in inflamed tissue as evaluated by microdialysis probes. Clin Pharmacol Ther 1990;48: [78] Hargreaves KM, Dionne RA, Mueller GP, Goldstein DS, Dubner R. Naloxone, fentanyl, and diazepam modify plasma beta-endorphin levels during surgery. Clin Pharmacol Ther 1986;40: [79] Harriott AM, Dessem D, Gold MS. Inflammation increases the excitability of masseter muscle afferents. Neuroscience 2006;141: [80] Harriott AM, Gold MS. Serotonin type 1D receptors (5HTR) are differentially distributed in nerve fibres innervating craniofacial tissues. Cephalalgia 2008;28: [81] Harriott AM, Gold MS. Contribution of primary afferent channels to neuropathic pain. Curr Pain Headache Rep 2009;13: [82] Harriott AM, Gold MS. Electrophysiological properties of dural afferents in the absence and presence of inflammatory mediators. J Neurophysiol 2009;101: [83] Henry MA, Luo S, Foley BD, Rzasa RS, Johnson LR, Levinson SR. Sodium channel expression and localization at demyelinated sites in painful human dental pulp. J Pain 2009;10: [84] Hokfelt TS, Ceccatelli L, Gustafsson AL, Hulting V, Verge M, Villar X J, Xu ZQ, Xu Z, Wiesenfeld-Hallin, Zhang X. Plasticity of NO synthase expression in the nervous and endocrine systems. Neuropharmacology 1994;33: [85] Hollins M, Harper D, Gallagher S, Owings EW, Lim PF, Miller V, Siddiqi MQ, Maixner W. Perceived intensity and unpleasantness of cutaneous and auditory stimuli: an evaluation of the generalized hypervigilance hypothesis. Pain 2009;141: [86] Hussain A, Stiles MA, Oshinsky ML. Pain remapping in migraine: a novel characteristic following trigeminal nerve injury. Headache 2010;50: [87] Inoue M, Fujii Y, Furukawa K, Okada M, Okumura K, Hayakawa T, Furukawa K, Sugiura Y. Refractory skin injury in complex knock-out mice expressing only the GM3 ganglioside. J Biol Chem 2002;277: [88] Jannetta PJ. Neurovascular compression in cranial nerve and systemic disease. Ann Surg 1980;192: [89] Jeske NA, Patwardhan AM, Gamper N, Price TJ, Akopian AN, Hargreaves KM. Cannabinoid WIN 55, regulates TRPV1 phosphorylation in sensory neurons. J Biol Chem 2006;281:

28 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 S31 [90] Jimenez-Andrade JM, Mantyh WG, Bloom AP, Xu H, Ferng AS, Dussor G, Vanderah TW, Mantyh PW. A phenotypically restricted set of primary afferent nerve fibers innervate the bone versus skin: therapeutic opportunity for treating skeletal pain. Bone 2010;46: [91] John MT, LeResche L, Koepsell TD, Hujoel P, Miglioretti DL, Micheelis W. Oral health-related quality of life in Germany. Eur J Oral Sci 2003;111: [92] John MT, Miglioretti DL, LeResche L, Von Korff M, Critchlow CW. Widespread pain as a risk factor for dysfunctional temporomandibular disorder pain. Pain 2003;102: [93] Katusic S, Beard CM, Bergstralh E, Kurland LT. Incidence and clinical features of trigeminal neuralgia, Rochester, Minnesota, Ann Neurol 1990;27: [94] Khan AA, Owatz CB, Schindler WG, Schwartz SA, Keiser K, Hargreaves KM. Measurement of mechanical allodynia and local anesthetic efficacy in patients with irreversible pulpitis and acute periradicular periodontitis. J Endod 2007;33: [95] Kim H, Lee H, Rowan J, Brahim J, Dionne RA. Genetic polymorphisms in monoamine neurotransmitter systems show only weak association with acute post-surgical pain in humans. Mol Pain 2006;2:24. [96] Kim H, Ramsay E, Lee H, Wahl S, Dionne RA. Genome-wide association study of acute post-surgical pain in humans. Pharmacogenomics 2009;10: [97] Kroner-Herwig B. Chronic pain syndromes and their treatment by psychological interventions. Curr Opin Psychiatry 2009;22: [98] Lee Y-S, Kim H, Wu T-X, Wang X-M, Dionne RA. Genetically mediated interindividual variation in analgesic responses to cyclooxygenase inhibitory drugs. Clin Pharmacol Ther 2006;79: [99] LeResche L. Epidemiology of temporomandibular disorders: implications for the investigation of etiologic factors. Crit Rev Oral Biol Med 1997;8: [100] LeResche L, Mancl LA, Drangsholt MT, Huang G, Von Korff M. Predictors of onset of facial pain and temporomandibular disorders in early adolescence. Pain 2007;129: [101] LeResche L, Mancl LA, Drangsholt MT, Saunders K, Korff MV. Relationship of pain and symptoms to pubertal development in adolescents. Pain 2005;118: [102] LeResche L, Mancl L, Sherman JJ, Gandara B, Dworkin SF. Changes in temporomandibular pain and other symptoms across the menstrual cycle. Pain 2003;106: [103] LeResche L, Saunders K, Von Korff MR, Barlow W, Dworkin SF. Use of exogenous hormones and risk of temporomandibular disorder pain. Pain 1997;69: [104] Levine JD, Gordon NC, Fields HL. The mechanism of placebo analgesia. Lancet 1978;2: [105] Levine JD, Gordon NC, Jones RT, Fields HL. The narcotic antagonist naloxone enhances clinical pain. Nature 1978;272: [106] Li YQ, Li H, Wei J, Qu L, Wu LA. Expression changes of K + -Cl co-transporter 2 and Na + -K + -Cl co-transporter1 in mouse trigeminal subnucleus caudalis following pulpal inflammation. Brain Res Bull 2010;81: [107] Lim PF, Smith S, Bhalang K, Slade GD, Maixner W. Development of temporomandibular disorders is associated with greater bodily pain experience. Clin J Pain 2010;26: [108] Lipton JA, Ship JA, Larach-Robinson D. Estimated prevalence and distribution of reported orofacial pain in the United States. J Am Dent Assoc 1994;124: [109] Lobb WK, Zakariasen KL, McGrath PJ. Endodontic treatment outcomes: do patients perceive problems? J Am Dent Assoc 1996;127: [110] Luo S, Perry GM, Levinson SR, Henry MA. Pulpitis increases the proportion of atypical nodes of Ranvier in human dental pulp axons without a change in Na v 1.6 sodium channel expression. Neuroscience 2010;169: [111] Macfarlane TV, Blinkhorn AS, Craven R, Zakrzewska JM, Atkin P, Escudier MP, Rooney CA, Aggarwal V, Macfarlane GJ. Can one predict the likely specific orofacial pain syndrome from a self-completed questionnaire? Pain 2004;111: [112] Macfarlane TV, Blinkhorn AS, Davies RM, Ryan P, Worthington HV, Macfarlane GJ. Orofacial pain: just another chronic pain? Results from a population-based survey. Pain 2002;99: [113] Malinow K, Yannakakis GD, Glusman SM, Edlow DW, Griffin J, Pestronk A, Powell DL, Ramsey-Goldman R, Eidelman BH, Medsger TA, Jr. Subacute sensory neuronopathy secondary to dorsal root ganglionitis in primary Sjögren s syndrome. Ann Neurol 1986;20: [114] Matthews B. The mechanisms of pain from dentine and pulp. Br Dent J 1976;140: [115] Mehlisch DR, Aspley S, Daniels SE, Southerden KA, Christensen KS. A singletablet fixed-dose combination of racemic ibuprofen/paracetamol in the management of moderate to severe postoperative dental pain in adult and adolescent patients: a multicenter, two-stage, randomized, double-blind, parallel-group, placebo-controlled, factorial study. Clin Ther 2010;32: [116] Melzack R. The McGill Pain Questionnaire: major properties and scoring methods. Pain 1975;1: [117] Melzack R, Jeans ME, Stratford JG, Monks RC. Ice massage and transcutaneous electrical stimulation: comparison of treatment for low-back pain. Pain 1980;9: [118] Miller JP, Acar F, Burchiel KJ. Classification of trigeminal neuralgia: clinical, therapeutic, and prognostic implications in a series of 144 patients undergoing microvascular decompression. J Neurosurg 2009;111: [119] Morgan CR, Bird EV, Robinson PP, Boissonade FM. TRPA1 expression in human lingual nerve neuromas in patients with and without symptoms of dysaesthesia. Neurosci Lett 2009;465: [120] Moskowitz MA, Macfarlane R. Neurovascular and molecular mechanisms in migraine headaches. Cerebrovasc Brain Metab Rev 1993;5: [121] Nackley AG, Shabalina SA, Tchivileva IE, Satterfield K, Korchynskyi O, Makarov SS, Maixner W, Diatchenko L. Human catechol-omethyltransferase haplotypes modulate protein expression by altering mrna secondary structure. Science 2006;314: [122] Nauta HJ, Wehman JC, Koliatsos VE, Terrell MA, Chung K. Intraventricular infusion of nerve growth factor as the cause of sympathetic fiber sprouting in sensory ganglia. J Neurosurg 1999;91: [123] Ni Riordain R, Moloney E, O Sullivan K, McCreary C. Burning mouth syndrome and oral health-related quality of life: is there a change over time? Oral Dis 2010;16: [124] Oh IH, Choi SK, Park BJ, Kim TS, Rhee BA, Lim YJ. The treatment outcome of elderly patients with idiopathic trigeminal neuralgia: micro-vascular decompression versus gamma knife radiosurgery. J Korean Neurosurg Soc 2008;44: [125] Okada-Ogawa A, Suzuki I, Sessle BJ, Chiang C-Y, Salter MW, Dostrovsky JO, Tsuboi Y, Kondo M, Kitagawa J, Kobayashi A, Noma N, Imamura Y, Iwata K. Astroglia in medullary dorsal horn (trigeminal spinal subnucleus caudalis) are involved in trigeminal neuropathic pain mechanisms. J Neurosci 2009;29: [126] Okamoto K, Bereiter DF, Thompson R, Tashiro A, Bereiter DA. Estradiol replacement modifies c-fos expression at the spinomedullary junction evoked by temporomandibular joint stimulation in ovariectomized female rats. Neuroscience 2008;156: [127] Owatz CB, Khan AA, Schindler WG, Schwartz SA, Keiser K, Hargreaves KM. The incidence of mechanical allodynia in patients with irreversible pulpitis. J Endod 2007;33: [128] Pajot J, Ressot C, Ngom I, Woda A. Gonadectomy induces site-specific differences in nociception in rats. Pain 2003;104: [129] Park S-S, Lee M-K, Kim J-W, Jung J-Y, Kim I-S, Ghang C-G. Percutaneous balloon compression of trigeminal ganglion for the treatment of idiopathic trigeminal neuralgia: experience in 50 patients. J Korean Neurosurg Soc 2008;43: [130] Patwardhan AM, Akopian AN, Ruparel NB, Diogenes A, Weintraub ST, Uhlson C, Murphy RC, Hargreaves KM. Heat generates oxidized linoleic acid metabolites that activate TRPV1 and produce pain in rodents. J Clin Invest 2010;120: [131] Patwardhan AM, Berg KA, Akopain AN, Jeske NA, Gamper N, Clarke WP, Hargreaves KM. Bradykinin-induced functional competence and trafficking of the delta-opioid receptor in trigeminal nociceptors. J Neurosci 2005;25: [132] Patwardhan AM, Diogenes A, Berg KA, Fehrenbacher JC, Clarke WP, Akopian AN, Hargreaves KM. PAR-2 agonists activate trigeminal nociceptors and induce functional competence in the delta opioid receptor. Pain 2006;125: [133] Patwardhan AM, Jeske NA, Price TJ, Gamper N, Akopian AN, Hargreaves KM. The cannabinoid WIN 55, inhibits transient receptor potential vanilloid 1 (TRPV1) and evokes peripheral antihyperalgesia via calcineurin. Proc Natl Acad Sci USA 2006;103: [134] Patwardhan AM, Scotland PE, Akopian AN, Hargreaves KM. Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia. Proc Natl Acad Sci USA 2009;106: [135] Pau AK, Croucher R, Marcenes W. Prevalence estimates and associated factors for dental pain: a review. Oral Health Prev Dent 2003;1: [136] Pepose JS, Foos RY, Stevens JG. Herpes simplex virus immediate-early protein ICP4 in murine models of latency. Graefes Arch Clin Exp Ophthalmol 1986;224: [137] Polycarpou N, Ng YL, Canavan D, Moles DR, Gulabivala K. Prevalence of persistent pain after endodontic treatment and factors affecting its occurrence in cases with complete radiographic healing. Int Endod J 2005;38: [138] Portenoy RK, Ugarte C, Fuller I, Haas G. Population-based survey of pain in the United States: differences among white, African American, and Hispanic subjects. J Pain 2004;5: [139] Price TJ, Helesic G, Parghi D, Hargreaves KM, Flores CM. The neuronal distribution of cannabinoid receptor type 1 in the trigeminal ganglion of the rat. Neuroscience 2003;120: [140] Putzki N, Pfriem A, Limmroth V, Yaldizli O, Tettenborn B, Diener HC, Katsarava Z. Prevalence of migraine, tension-type headache and trigeminal neuralgia in multiple sclerosis. Eur J Neurol 2009;16: [141] Quang PN, Schmidt BL. Endothelin-A receptor antagonism attenuates carcinoma-induced pain through opioids in mice. J Pain 2010;11: [142] Robert C, Wilson CS, Donnadieu S, Gaudy JF, Arreto CD. Bibliometric analysis of the scientific literature on pain research: a 2006 study. Pain 2008;138: [143] Robinson PP, Smith KG. Lingual nerve damage during lower third molar removal: a comparison of two surgical methods. Br Dent J 1996;180: [144] Robinson JP, Willars GB, Tomlinson DR, Keen P. Axonal transport and tissue contents of substance P in rats with long-term streptozotocin-diabetes. Effects of the aldose reductase inhibitor statil. Brain Res 1987;426:

29 S32 K.M. Hargreaves / PAIN Ò 152 (2011) S25 S32 [145] Rowan MP, Ruparel NB, Patwardhan AM, Berg KA, Clarke WP, Hargreaves KM. Peripheral delta opioid receptors require priming for functional competence in vivo. Eur J Pharmacol 2009;602: [146] Ruparel NB, Patwardhan AM, Akopian AN, Hargreaves KM. Homologous and heterologous desensitization of capsaicin and mustard oil responses utilize different cellular pathways in nociceptors. Pain 2008;135: [147] Salas MM, Hargreaves KM, Akopian AN. TRPA1-mediated responses in trigeminal sensory neurons: interaction between TRPA1 and TRPV1. Eur J Neurosci 2009;29: [148] Scaravilli F. Reduced substance P in hereditary sensory neuropathy in the mf rat. Brain Res 1983;263: [149] Sessle BJ, Hu JW. Mechanisms of pain arising from articular tissues. Can J Physiol Pharmacol 1991;69: [150] Seymour RA, Charlton JE, Phillips ME. An evaluation of dental pain using visual analogue scales and the Mcgill Pain Questionnaire. J Oral Maxillofac Surg 1983;41: [151] Sigurdsson A, Maixner W. Effects of experimental and clinical noxious counterirritants on pain perception. Pain 1994;57: [152] Simpson MT, MacLaurin JG, Xu D, Ferguson KL, Vanderluit JL, Davoli MA, Roy S, Nicholson DW, Robertson GS, Park DS, Slack RS. Caspase 3 deficiency rescues peripheral nervous system defect in retinoblastoma nullizygous mice. J Neurosci 2001;21: [153] Siqueira SR, Alves B, Malpartida HM, Teixeira MJ, Siqueira JT. Abnormal expression of voltage-gated sodium channels Nav1.7, Nav1.3 and Nav1.8 in trigeminal neuralgia. Neuroscience 2009;164: [154] Slade GD, Diatchenko L, Bhalang K, Sigurdsson A, Fillingim RB, Belfer I, Max MB, Goldman D, Maixner W. Influence of psychological factors on risk of temporomandibular disorders. J Dent Res 2007;86: [155] Smeyne R J, Klein R, Schnapp A, Long LK, Bryant S, Lewin A, Lira SA, Barbacid M. Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene. Nature 1994;368: [156] Smith TJ, Morrison LA, Leib DA. Pathogenesis of herpes simplex virus type 2 virion host shutoff (vhs) mutants. J Virol 2002;76: [157] Spivack JG, Fareed MU, Valyi-Nagy T, Nash TC, O Keefe JS, Gesser RM, McKie EA, MacLean AR, Fraser NW, Brown SM. Replication, establishment of latent infection, expression of the latency-associated transcripts and explant reactivation of herpes simplex virus type 1 gamma 34.5 mutants in a mouse eye model. J Gen Virol 1995;76: [158] Stephenson J, Byers MR. GFAP immunoreactivity in trigeminal ganglion satellite cells after tooth injury in rats. Expt Neurol 1995;131: [159] Sterman AB, Schaumburg HH, Asbury AK. The acute sensory neuronopathy syndrome: a distinct clinical entity. Ann Neurol 1980;7: [160] Stohler CS, Kowalski CJ. Spatial and temporal summation of sensory and affective dimensions of deep somatic pain. Pain 1999;79: [161] Sweet WH. Deafferentation pain after posterior rhizotomy, trauma to a limb, and herpes zoster. Neurosurgery 1984;15: [162] Swift JQ, Garry MG, Roszkowski MT, Hargreaves KM. Effect of flurbiprofen on tissue levels of immunoreactive bradykinin and acute postoperative pain. J Oral Maxillofac Surg 1993;51:112 6 [discussion 116 7]. [163] Tacconi L, Miles JB. Bilateral trigeminal neuralgia: a therapeutic dilemma. Br J Neurosurg 2000;14:33 9. [164] Tahmoush AJ. Causalgia: redefinition as a clinical pain syndrome. Pain 1981;10: [165] Tal M, Devor M. Ectopic discharge in injured nerves: comparison of trigeminal and somatic afferents. Brain Res 1992;579: [166] Tanaka T, Sakamoto E, Shiiba S, Oda M, Kito S, Wakasugi-Sato N, Matsumoto- Takeda S, Imamura Y, Nakanishi O, Morimoto Y. Relationship between the curative effects of carbamazepine administration and the neurovascular compression volume of the trigeminal nerve measured using magnetic resonance cisternography. Clin J Pain 2009;25: [167] Tarricone R, Aguzzi G, Musi F, Fariselli L, Casasco A. Cost-effectiveness analysis for trigeminal neuralgia: cyberknife vs microvascular decompression. Neuropsychiatr Dis Treat 2008;4: [168] Tashiro A, Okamoto K, Bereiter DA. Chronic inflammation and estradiol interact through MAPK activation to affect TMJ nociceptive processing by trigeminal caudalis neurons. Neuroscience 2009;164: [169] Tchivileva IE, Lim PF, Smith SB, Slade GD, Diatchenko L, McLean SA, Maixner W. Effect of catechol-o-methyltransferase polymorphism on response to propranolol therapy in chronic musculoskeletal pain: a randomized, doubleblind, placebo-controlled, crossover pilot study. Pharmacogenet Genomics 2010;20: [170] Thackray AM, Field HJ. Differential effects of famciclovir and valaciclovir on the pathogenesis of herpes simplex virus in a murine infection model including reactivation from latency. J Infect Dis 1996;173: [171] Tsuzuki K, Kondo E, Fukuoka T, Yi D, Tsujino H, Sakagami M, Noguchi K. Differential regulation of P2X(3) mrna expression by peripheral nerve injury in intact and injured neurons in the rat sensory ganglia. Pain 2001;91: [172] U.S. Department of Health and Human Services. Oral health in America: a report of the Surgeon General. Rockville, MD: U.S. Department of Health and Human Services. National Institute of Dental and Craniofacial Research, National Institutes of Health; [173] Valmaseda-Castellon E, Berini-Aytes L, Gay-Escoda C. Lingual nerve damage after third lower molar surgical extraction. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2000;90: [174] Vargas CM, Macek MD, Marcus SE. Sociodemographic correlates of tooth pain among adults: United States, Pain 2000;85: [175] Vaughn AH, Gold MS. Ionic mechanisms underlying inflammatory mediatorinduced sensitization of dural afferents. J Neurosci 2010;30: [176] Vickers ER, Cousins MJ, Woodhouse A. Pain description and severity of chronic orofacial pain conditions. Aust Dent J 1998;43: [177] Vos BP, Strassman AM, Maciewicz RJ. Behavioral evidence of trigeminal neuropathic pain following chronic constriction injury to the rat s infraorbital nerve. J Neurosci 1994;14: [178] Warren CA, Mok L, Gordon S, Fouad AF, Gold MS. Quantification of neural protein in extirpated tooth pulp. J Endod 2008;34:7 10. [179] White TM, Gilden DH, Mahalingam R. An animal model of varicella virus infection. Brain Pathol 2001;11: [180] Wilkins RH. Neurovascular compression syndromes. Neurol Clin 1985;3: [181] Woodham P, Anderson P, Nadim W, Trurmaine M. Satellite cells surrounding axotomized rat dorsal root ganglion cells increase expression of GFAP-like proetin. Neurosci Lett 1989;98:8 12. [182] Wu YW, Bi YP, Kou XX, Xu W, Ma LQ, Wang KW, Gan YH, Ma XC. 17-Betaestradiol enhanced allodynia of inflammatory temporomandibular joint through upregulation of hippocampal TRPV1 in ovariectomized rats. J Neurosci 2010;30: [183] Youle M, Osio M. A double-blind, parallel-group, placebo-controlled, multicentre study of acetyl L-carnitine in the symptomatic treatment of antiretroviral toxic neuropathy in patients with HIV-1 infection. HIV Med 2007;8: [184] Zakrzewska JM, Jassim S, Bulman JS. A prospective, longitudinal study on patients with trigeminal neuralgia who underwent radiofrequency thermocoagulation of the Gasserian ganglion. Pain 1999;79:51 8. [185] Zhang X, Ji RR, Arvidsson J, Lundberg JM, Bartfai T, Bedecs K, Hokfelt T. Expression of peptides, nitric oxide synthase and NPY receptor in trigeminal and nodose ganglia after nerve lesions. Exp Brain Res 1996;111: [186] Zhang JM, Song X, LaMotte R. Enhanced excitability of sensory neurons in rats with cutaneous hyperalgesia produced by chronic compression of the dorsal root ganglion. J Neurophysiol 2002;82:

30 PAIN Ò 152 (2011) S33 S40 Review Pathophysiology of postoperative pain Timothy J. Brennan Departments of Anesthesia and Pharmacology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article. 1. Introduction 1.1. Clinical perspective Acute postoperative pain remains a significant medical problem. Patients undergoing outpatient ambulatory surgery have clinically significant postoperative pain, even when administration of oral opioids and nonopioid adjuncts are optimized [1]. Regional analgesic techniques improve pain control, except their use is limited to a minority of all surgical patients [8,31]. For those patients undergoing major surgical procedures, ongoing pain or pain at rest, and pain during activities are important clinical symptoms. Pain at rest is usually moderate; the average visual analogue pain scale (VAS) score is 3 to 4 of 10 during the first 2 to 3 days after surgery [24]. These pain scores occur even when parenteral treatments are administered. Usually, pain at rest resolves within the first week after surgery (see schematic, Fig. 1). Pain with activities, such as coughing or walking, is severe during the first 2 to 3 days; the average VAS score can be as great as 7 to 8. Pain with activities is moderate or severe for many days and even weeks later. Functional capability is limited during this period as well; thus, pain can be moderate, and the activities such as ability to cough or walking distance to evoke this pain are reduced [16,36]. Greater opioid dosing to further reduce pain is limited by side effects such as nausea, vomiting, ileus, respiratory depression, and sedation [8]. Clinically, our goal is to advance simple, safe, effective therapies that will greatly reduce postoperative pain. In the last 10 to 15 years, gabapentinoids and cyclooxygenase-2 inhibitors have undergone extensive trials, and regional anesthesia is advancing in acceptance [9]. In general, we have had difficulty making major improvements in the overall treatment of clinical postoperative pain for the majority of patients Scientific perspective Currently, we recognize that the etiology and treatment of pain produced by surgery is different than other clinical pain conditions such as rheumatoid arthritis, fibromyalgia, or acute herpes zoster Address: Department of Anesthesia, 200 Hawkins Drive, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA. Tel.: ; fax: address: tim-brennan@uiowa.edu [6,23]. Many pain models may be useful for mechanistic studies but less valuable for developing novel targets to treat particular clinical pain conditions. To advance our treatment of acute postoperative pain, we must recognize that many preclinical pain models, such as antigen-specific inflammation or receptor-specific chemical stimuli (ie, formalin or capsaicin), do not necessarily translate well to incisional pain mechanisms [15,44,46,47]. Theoretically, if the etiology and pathophysiology of the clinical condition are mimicked by the experimental model, analgesic target discovery may be improved. A second goal for preclinical rodent pain research is to make the end points for nociceptive testing have proximate translatability to clinical pain and analgesia [34]. Others have suggested that plasma levels of drugs for antinociception in animals and plasma concentrations for analgesia in human subjects be compared to improve the translatability of the models and the behavior in rodents [34]. Limitations of pain models and behaviors may be one of several factors contributing to our limited success in developing new treatments for acute postoperative pain [20,32]. Because there has been considerable debate regarding the clinical utility of rodent pain models, we and others undertook studies on the etiology of nociception caused by incisions [2,12,21,26, 28,39]. Perhaps improving our understanding of the mechanisms for postoperative pain could increase the chances of developing new treatments for patients. It is our hope that research on incisional pain mechanisms and relating these preclinical results to patient symptoms has advanced our understanding of animal pain models; incision-induced, painrelated behaviors in animal models; and postoperative pain mechanisms in patients. The long-term goal of postoperative pain research and acute incisional pain studies is for patients to undergo painless or nearly painless surgery. 2. Preclincal studies 2.1. Rat plantar incision Several years ago, it was noted that few studies used incisions to understand mechanisms of pain caused by surgery. At the time of our model development, many pain behaviors used hind paw injections into the glabrous skin and nociception was measured using behaviors associated with hind paw withdrawal [14]. We undertook experiments using hind paw incision to use similar /$36.00 Ó 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved. doi: /j.pain

31 VAS S34 T.J. Brennan / PAIN Ò 152 (2011) S33 S cough 1-minute scoring period over 1 hour. Guarding is increased after incision and gradually returns towards preincision values over the next 2 to 3 days. The rapid resolution of guarding pain after plantar incision, although a robust mechanical and heat hyperalgesia are still evident, suggests different mechanisms. We were interested in the relationship of guarding behavior to spontaneous activity (SA) in nociceptive pathways and postoperative pain in patients. If SA in part signals nonevoked, guarding pain in nociceptors and dorsal horn neurons, both measures, guarding and SA, may be in part translatable to pain at rest in patients after surgery. 0 behaviors and to compare these responses to other models [5]. Other preclinical surgical models were developed. For plantar incision, we use general anesthesia. The hind paw is sterilized, and the glabrous skin is incised usually through the skin and underlying plantar fascia, which are in close apposition in vivo (Fig. 2A D). After incising to the fascia, an underlying flexor digitorum brevis muscle is encountered. This muscle contracts the lateral 4 digits of the hind paw. This underlying flexor muscle is elevated, divided, and retracted. Then the skin overlying the muscle is closed with sutures. The rat is allowed to recover from general anesthesia, and pain behaviors can be tested as early as 1 hour after incision. In patients who undergo surgical procedures that access deep tissues, skin is incised and underlying muscles are divided and retracted so that inflamed organs or cancerous tumors can be removed. Therefore, the somatic injury after plantar incision has some similarities to the somatic injury of some patients undergoing surgery Evoked pain behaviors A variety of pain-related, nociceptive behaviors can be measured after plantar incision [45]. Reduced heat withdrawal latency is greatest the day of incision and sustained for approximately 5 days, and is completely resolved by 7 to 10 days after the procedure. Withdrawal threshold to punctate monofilament application adjacent to the incision for mechanical testing is also markedly decreased, and this response is sustained for 5 to 10 days. Both primary mechanical hyperalgesia and primary heat hyperalgesia are present in patients after surgery [22,37]. Finally, other pain behaviors have been measured. These include Randall-Selitto mechanical withdrawal threshold [40], weight bearing [40], and secondary mechanical withdrawal threshold [45] Guarding Pre Op Days After Surgery Fig. 1. Schematic of postoperative pain after major surgery in patients with optimized parenteral opioid analgesia. Top line is pain with cough; bottom line is pain at rest. VAS, visual analogue scale. Immediately after plantar incision, there is a small amount of spontaneous pain behavior, such as licking of the incised hind paw. This usually resolves 30 minutes to 1 hour after emergence from anesthesia [5]. We have observed that rats guard the incised hind paw early in the postoperative period. We have measured nonevoked pain behavior using a cumulative pain score that examines the position in which both paws are placed on a plastic mesh floor. A cumulative pain score is obtained depending on the position in which the paw is observed during the majority of the rest 3. In vivo neurophysiology experiments 3.1. Spontaneous activity in nociceptive pathways after incision A key to translational pain research is to identity sensitized nociceptors and dorsal horn neurons that encode enhanced nociception so that targets transmitting these amplified signals can be evaluated for therapeutic potential. Ongoing activity or SA, one aspect of sensitization, is a stimulus-independent measurement of sensitization [23,33,35]. We have undertaken a series of electrophysiological studies in rats that had undergone plantar incision 1 day earlier and examined primary afferent and dorsal horn neuron activity and compared SA with unincised, sham-operated animals. Most studies in our laboratory include not only measurements of ongoing SA, but also heat, mechanical, and in some cases chemical sensitivity. SA is perhaps the most robust form of sensitization produced in postinjury states because no stimulus is required [19]. This review focuses on the generation of or an increase in the magnitude of SA of nociceptors and dorsal horn neurons produced by incisions. One day after plantar incision, guarding behavior is usually significantly elevated [5]. We compared the SA of primary afferent nociceptors from rats that had undergone plantar incision [29], which included incisions in skin, fascia, and muscle 1 day earlier than those that had undergone sham operation (Fig. 3). The nociceptors studied from the incision group innervated, at least in part, the area of the incision. Thirty-nine afferent fibers were recorded in the sham-operated group, and 28 fibers were recorded in the plantar incision group on postoperative day 1. In the incision group, 11 of 28 nociceptors had SA; the average frequency was approximately 17 imp/s. In this study, the majority of spontaneously active Ad and C-fiber activity was >15 imp/s after incision. In the sham group, no afferents were spontaneously active. The origin of the SA was the incised tissue because local anesthetic infiltrated into the receptive field of the nociceptor in the hind paw eliminated the activity [29]. It was surprising that the rate of SA was quite high in some nociceptive fibers. We performed the corresponding experiment, but this time recording dorsal horn neurons [43]. We measured behavior in rats that had undergone skin, fascia, and muscle incision on postoperative day 1 and compared the behavior with that of rats that underwent a sham operation (Fig. 4). The mean guarding pain scores were 15 and 0 in the incised and sham-operated groups, respectively. Rats then underwent electrophysiology experiments, recordings of single dorsal horn neuron activity under general anesthesia. These dorsal horn neurons received input from afferent fibers innervating the glabrous skin of the hind paw and had a mechanoreceptive field that again included at least part of the area of the incision. SA was identified in 9 of 27 neurons in the control group and 16 of 26 neurons in the incision group. The average dorsal horn SA in the incised rats was 19 imp/s, and was 10 imp/s in the sham-operated group. In this series, both the proportion of neurons with SA and the magnitude of SA were greater in the 29

32 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 S35 neurons from the incised group. Bupivacaine injection into the skin and deep tissue of the hind paw reduced the level of SA in the incised group to the same level of that in the sham-operated group [43]. Bupivacaine did not affect the magnitude of SA in the sham group. In summary, although SA is intrinsic to some dorsal horn neurons, the proportion of neurons with SA and the amount of SA were both increased 1 day after incision. Altogether, increased spinal dorsal horn neuron activity remained largely dependent on ongoing primary afferent input on postoperative day 1 when guarding behavior was evident. The SA was quite high in dorsal horn neurons and in agreement with the magnitude of activity in primary afferent nociceptors in similar groups of animals (Fig. 3) Tissues contributing to guarding pain and SA of nociceptive pathways Three previous studies identified cutaneous nociceptors and examined SA before and after skin injury using scalpel blades or Fig. 2. Photographs of the different stages of the rat plantar incision. (A) A 1-cm longitudinal incision is made through the skin and fascia starting 0.5 cm from the proximal edge of the heel and extending toward the distal aspect of the paw. (B and C) The underlying flexor muscle is elevated and also incised longitudinally. The muscle is split and dissected longitudinally. (D) After hemostasis, the wound is apposed with 2 mattress sutures of 5 0 nylon. 30

33 S36 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 A B Activity (volts) ms C D percentage of afferents (%) /39 * 11/28 mean rate (imp/s) * control incision control incision (n=11) Fig. 3. Spontaneous activity in nociceptors 1 day after plantar incision [29]. (A) Schematic of in vivo recording from nociceptors from rats that underwent hind paw incision. (B) Example of spontaneous action potentials recorded from a nociceptor in a rat that underwent plantar incision. (C) Percentage of nociceptors with spontaneous activity in the control group that underwent a sham operation and group that underwent skin, fascia, and muscle incision [29,42]. (D) Mean rate of spontaneous activity of nociceptors that underwent skin, fascia and muscle incision [29,42]. needles. In 2 separate studies, investigators used needles to penetrate the receptive fields of both A-delta and C-fiber nociceptors recorded from cutaneous nerves in the cat [3,7]. Activation during needle penetrations occurred; however, no sustained SA was generated after the injury. Finally, Hamalainen et al. [13] recorded afferent fibers from the tibial nerve innervating the hind paw of the rat. Activity was generated during incision and other forms of sensitization were evident, yet no nociceptors developed ongoing, sustained SA after incision. It was surprising that nociceptors could not be activated immediately after incision because dorsal horn neurons generate some sustained SA during and after the same incisions [30,38,46]. Dorsal horn neurons receive convergent input from a variety of tissues, whereas nociceptors innervate specific tissues. Perhaps by selecting cutaneous nociceptors and incising their receptive fields, these cutaneous nociceptors did not generate SA after incision. We hypothesized that deep tissue incision that included fascia and muscle would generate SA in nociceptive pathways and produce unprovoked guarding pain, but a skin-only incision would not. We undertook a series of experiments incising skin only, skin, fascia and muscle (deep tissue), or sham (no operation) surgery. Guarding behavior was measured in separate groups of rats after sham, skin, or skin plus deep tissue incision 1 day later. A fourth group was studied 7 days after skin plus deep tissue incision, when pain behaviors generally resolve. These same groups then underwent in vivo recording of single-fiber nociceptors innervating the area of the incision (Fig. 5A C). Compared with the sham control group, skin incision induced a small amount of guarding on the day of incision only, whereas skin plus deep tissue incision caused guarding for 5 days after incision and this behavior completely resolved on postoperative day 7 [41,42]. On postoperative day 1, skin incision (18%) produced a similar prevalence of SA in nociceptors as the sham-operated group (13%), whereas skin plus deep tissue incision generated a greater prevalence of SA in nociceptors (61%); the rate of SA also tended to be greater after skin plus deep tissue incision (10 ± 3 imp/s) versus the control group (6 ± 6) and skin incision group (6 ± 3 imp/s). Seven days after skin plus deep tissue incision, when pain behaviors had resolved, the prevalence of SA (14%) and the amount of SA (0.3 ± 0.1) were similar to that in the sham group [42]. Incision of skin was not sufficient to produce guarding pain behavior, but incision that included skin fascia and muscle was sufficient. In addition, skin incision was sufficient to produce the typical reduced heat withdrawal latency and reduced mechanical withdrawal threshold on postoperative day 1 [41,42]. A comparable series of experiments in 4 similar groups of rats was undertaken, except in these 4 groups (Fig. 5D F), dorsal horn neurons were recorded on postoperative day 1 [41]. Pain behaviors again showed the importance of deep muscle incision on the generation of guarding, but normal heat and mechanical responses after skin-only incision. On postoperative day 1, skin incision (53%) produced a similar prevalence of SA in dorsal horn neurons as in the sham-operated group (36%), whereas skin plus deep tissue incision generated a greater prevalence of SA in dorsal horn neurons (78%); the rate of SA also tended to be greater after skin plus deep tissue incision (14 ± 3 imp/s) versus the control group (6 ± 2) and skin incision group (9 ± 2 imp/s). Seven days after skin plus deep tissue incision, when pain behaviors had resolved, the prevalence of SA (30%) and the amount of SA (6 ± 2) were similar to that in the sham group. Bupivacaine infiltration into the incision 31

34 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 S37 A B 1.5 Activity (volts) C D E cumulative pain score *** percentage of neurons (%) /27 * 16/26 mean rate (imp/s) * control (n=17) incision (n=17) control incision control (n=9) incision (n=16) Fig. 4. Spontaneous activity in dorsal horn neurons 1 day after plantar incision [43]. (A) Schematic of dorsal horn neuron recording. (B) Example of spontaneous action potentials recorded from dorsal horn neuron in a rat that underwent plantar incision. Reproduced from PAIN 144: with permission of the International Association for the Study of Pain Ò (IASP Ò ). The figure may not be reproduced for any other purpose without permission. (C) Guarding pain score in rats that underwent a sham (control) operation and a group that underwent skin, fascia, and muscle incision. (D) Percentage of dorsal horn neurons with spontaneous activity in the control group and group that underwent skin, fascia, and muscle incision. (E) Mean rate of spontaneous activity of dorsal horn neurons in the control and incision groups. C and D adapted with permission [43, p. 823 and 824]. A B C percentage of afferents (%) /23 ** 4/22 * 14/23 ** 3/22 mean rate (imps/s) control skin skin+deep POD1 skin+deep POD7 control (n=3) skin (n=4) POD1 skin+deep (n=14) skin+deep (n=3) POD7 D E percentage of neurons (%) ** *** 25/32 16/30 10/28 10/34 control skin skin+deep skin+deep POD1 POD7 F mean rate (imps/s) control (n=10) skin (n=16) POD1 skin+deep (n=25) skin+deep (n=10) POD7 Fig. 5. Spontaneous activity in nociceptors and dorsal horn neurons. (A C) Nociceptor recordings in rats that underwent a sham (control) operation; a skin incision; a skin, fascia, and muscle incision; and a group 7 days after skin, fascia, and muscle incision. Percentage of nociceptors with spontaneous activity (B) and average spontaneous activity (C). B and C reprinted with permission [42]. (D F) Dorsal horn neuron recordings in rats that underwent a sham (control) operation; a skin incision; a skin, fascia, and muscle incision; and a group 7 days after skin, fascia, and muscle incision. Percentage of dorsal horn neurons with spontaneous activity (E) and average spontaneous activity (F). E and F adapted and reprinted with permission [41]. E and F have been reproduced with permission from the International Association for the Study of Pain Ò (IASP Ò ). The figure may not be reproduced for any other purpose without permission. 32

35 S38 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 decreased SA in the skin plus deep tissue incision group to the same level as in the sham-operated rats on postoperative day 1 [41]. These data showed that incised deep tissue rather than skin had a central role in the genesis of guarding behavior and SA in nociceptors and nociceptive transmitting dorsal horn neurons (Fig. 6). Nociceptor and dorsal horn SA was associated with guarding behavior after incision. Decreased heat withdrawal latency and decreased mechanical withdrawal threshold occurred with skin incision only. 4. Human studies 4.1. Forearm incision Data from human studies support these preclinical incisional pain studies [17,18]. First, when an incision was made in the forearms of human volunteers, pain diminished soon after the blade had been pulled out from the skin; spontaneous pain rapidly decreased and resolved within 30 minutes after incision. However, mechanical hyperalgesia was evident. The hyperalgesia was similar in magnitude and duration to the mechanical withdrawal thresholds in rats after plantar incision. Thus, forearm incision in volunteers has similarities to skin incision of the rat hind paw Clinical studies In support of this concept, 2 surgical approaches for a unilateral total hip arthroplasty, a minimally invasive approach and a conventional approach, were recently compared [11]. Both approaches used the same length of skin incision, 20 cm. The minimally invasive approach preserved the underlying muscles, whereas muscles were incised and divided in the conventional approach. The minimally invasive approach resulted in significantly less postoperative pain and opioid consumption than the conventional approach. In a complimentary study [27], 2 groups of patients underwent total hip arthroplasty with the same amount of deep tissue dissection through different lengths of skin incision (10 vs 20 cm). There was no difference in postoperative pain between groups when the same degree of deep tissue injury occurred. Together, these postoperative pain studies indicate that reducing the amount of deep tissue injury decreases pain at rest and opioid consumption, whereas varying the magnitude of the skin incision did not affect pain at rest or opioid use. 5. Implications for animal models, clinical disease states, and drug discovery In preclinical behavioral studies, there has been a significant emphasis on exaggerated withdrawal responses in models of pathologic pain states. This began with testing changes in heat withdrawal latency and subsequently punctate mechanical withdrawal thresholds. Both heat hyperalgesia [22] and decreased mechanical pain threshold [37] occur in surgical patients. Because there has been increasing awareness of the limitations of these evoked behavioral responses, the applicability of cutaneous hyperalgesia to patient postoperative pain symptoms may be limited. Fig. 6. Schematic for the development guarding pain and SA in the nociceptive pathways after plantar incision. (A) An incision in the skin only (epidermis and dermis layer) induces minimal SA in nociceptors and dorsal horn neurons, which receive predominately cutaneous input. (B) An incision including skin and deep tissue (fascia and muscle layer) results in sustained SA in muscle-innervating primary afferents and the dorsal horn neurons receiving input from muscle. Reprinted with permission [42]. 33

36 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 S39 The implications of our data are the following. Clinical studies with local anesthetic infiltration and nerve blockade indicate that preincision analgesia has little clinical benefit compared with administration of analgesic drugs later after surgery [25]. Basic science data indicate that early after surgery, primary afferent activation and peripheral sensitization are profound when patient s postoperative pain is greatest [29,41 43]. Central sensitization occurs early in the postoperative period, but its precise role in clinical acute pain is not clear [10]. Central sensitization likely contributes to referred pain and secondary hyperalgesia; perhaps it is related to chronic posttraumatic pain [4]. There is profound primary afferent and dorsal horn neuron activation 1 day after plantar incision [29,42]. Despite evidence that some nociceptors have ongoing activity P20 imp/s, no obvious licking, biting, or scratching of the hind paw is evident 1 day after plantar incision. These data suggest that behaviors resulting from ongoing activity in nociceptive pathways may be modest and difficult to detect behaviorally, but may represent very clinically significant acute pain. The demonstration of high ongoing activity in dorsal horn neurons and primary afferent fibers suggests that these neurophysiology experiments may detect activation of the nociceptive system when spontaneous nociceptive behaviors are limited. Using evoked stimuli to test the effect of an analgesic drug could fail to capture drugs effective against ongoing activity in nociceptive pathways, a correlate to human pain at rest. Thus, a useful analgesic drug may be overlooked when only evoked responses are tested. Tests that utilize mechanical and heat stimuli may largely evaluate cutaneous sensitivity after incision. This may apply to other models as well. Skin injury and skin testing may have limited clinical relevance and may in part have contributed to the limited discovery of new analgesic drugs. The skin is certainly an important model system, but clinical pathophysiologic relevance may be limited for postoperative patients. 6. Conclusions Clinical studies indicate that improvements in postoperative pain control will advance perioperative medicine. Models for pain and nociception continue to be refined and evaluated. For postoperative models, the behavioral responses after incision and neurophysiologic studies on activities of primary afferent and dorsal horn neurons indicate that different tissues have unique responses to incision. These studies hopefully will improve our understanding of incisional pain mechanisms and the behavioral aspects of activation and sensitization of nociceptive pathways. Conflict of interest statement The author has no conflict of interest to declare. References [1] Apfelbaum JL, Chen C, Mehta SS, Gan TJ. Postoperative pain experience: results from a national survey suggest postoperative pain continues to be undermanaged. Anesth Analg 2003;97: [2] Beitz AJ, Newman A, Shepard M, Ruggles T, Eikmeier L. A new rodent model of hind limb penetrating wound injury characterized by continuous primary and secondary hyperalgesia. J Pain 2004;5: [3] Bessou P, Perl ER. Response of cutaneous sensory units with unmyelinated fibers to noxious stimuli. J Neurophysiol 1969;32: [4] Brennan TJ, Kehlet H. Preventive analgesia to reduce wound hyperalgesia and persistent postsurgical pain: not an easy path. Anesthesiology 2005;103: [5] Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain 1996;64: [6] Brennan TJ, Zahn PK, Pogatzki-Zahn EM. Mechanisms of incisional pain. Anesthesiol Clin North Am 2005;23:1 20. [7] Burgess PR, Perl ER. Myelinated afferent fibres responding specifically to noxious stimulation of the skin. J Physiol 1967;190: [8] Dahl JB, Kehlet H. Postoperative pain and its management. In: McMahon SB, Koltzenburg M, editors. Wall and Melzack s textbook of pain. Amsterdam, The Netherlands: Elsevier; p [9] Dahl JB, Mathiesen O, Kehlet H. An expert opinion on postoperative pain management, with special reference to new developments. Expert Opin Pharmacother 2010;11: [10] Dirks J, Moiniche S, Hilsted KL, Dahl JB. Mechanisms of postoperative pain: clinical indications for a contribution of central neuronal sensitization. Anesthesiology 2002;97: [11] Dorr LD, Maheshwari AV, Long WT, Wan Z, Sirianni LE. Early pain relief and function after posterior minimally invasive and conventional total hip arthroplasty. A prospective, randomized, blinded study. J Bone Joint Surg Am 2007;89: [12] Duarte AM, Pospisilova E, Reilly E, Hamaya Y, Mujenda F, Strichartz GR. Reduction of post-incisional allodynia by subcutaneous bupivacaine: findings with a new model in the hairy skin of the rat. Anesthesiology 2005;103: [13] Hamalainen MM, Gebhart GF, Brennan TJ. Acute effect of an incision on mechanosensitive afferents in the plantar rat hind paw. J Neurophysiol 2002;87: [14] Hargreaves K, Dubner R, Brown F, Flores C, Joris J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1988;32: [15] Honore P, Wade CL, Zhong C, Harris RR, Wu C, Ghayur T, Iwakura Y, Decker MW, Faltynek C, Sullivan J, Jarvis MF. Interleukin-1alphabeta gene-deficient mice show reduced nociceptive sensitivity in models of inflammatory and neuropathic pain but not post-operative pain. Behav Brain Res 2006;167: [16] Ilfeld BM, Mariano ER, Girard PJ, Loland VJ, Meyer RS, Donovan JF, Pugh GA, Le LT, Sessler DI, Shuster JJ, Theriaque DW, Ball ST. A multicenter, randomized, triple-masked, placebo-controlled trial of the effect of ambulatory continuous femoral nerve blocks on discharge-readiness following total knee arthroplasty in patients on general orthopaedic wards. Pain 2010;150: [17] Kawamata M, Takahashi T, Kozuka Y, Nawa Y, Nishikawa K, Narimatsu E, Watanabe H, Namiki A. Experimental incision-induced pain in human skin: effects of systemic lidocaine on flare formation and hyperalgesia. Pain 2002;100: [18] Kawamata M, Watanabe H, Nishikawa K, Takahashi T, Kozuka Y, Kawamata T, Omote K, Namiki A. Different mechanisms of development and maintenance of experimental incision-induced hyperalgesia in human skin. Anesthesiology 2002;97: [19] Liang YF, Haake B, Reeh PW. Sustained sensitization and recruitment of rat cutaneous nociceptors by bradykinin and a novel theory of its excitatory action. J Physiol 2001;532: [20] Mao J. Translational pain research: achievements and challenges. J Pain 2009;10: [21] Martin TJ, Buechler NL, Kahn W, Crews JC, Eisenach JC. Effects of laparotomy on spontaneous exploratory activity and conditioned of operant responding in the rat a model for postoperative pain. Anesthesiology 2004;101: [22] Martinez V, Fletcher D, Bouhassira D, Sessler DI, Chauvin M. The evolution of primary hyperalgesia in orthopedic surgery: quantitative sensory testing and clinical evaluation before and after total knee arthroplasty. Anesth Analg 2007;105: [23] Meyer RA, Ringkamp M, Campbell JN, Raja SN. Peripheral mechanisms of cutaneous nociception. In: McMahon SB, Koltzenburg M, editors. Wall and Melzack s textbook of pain. Amsterdam, The Netherlands: Elsevier; p [24] Moiniche S, Dahl JB, Erichsen CJ, Jensen LM, Kehlet H. Time course of subjective pain ratings, and wound and leg tenderness after hysterectomy. Acta Anaesthesiol Scand 1997;41: [25] Moiniche S, Kehlet H, Dahl JB. A qualitative and quantitative systematic review of preemptive analgesia for postoperative pain relief: the role of timing of analgesia. Anesthesiology 2002;96: [26] Nara T, Saito S, Obata H, Goto F. A rat model of postthoracotomy pain: behavioural and spinal cord NK-1 receptor assessment. Can J Anaesth 2001;48: [27] Ogonda L, Wilson R, Archbold P, Lawlor M, Humphreys P, O Brien S, Beverland D. A minimal-incision technique in total hip arthroplasty does not improve early postoperative outcomes. A prospective, randomized, controlled trial. J Bone Joint Surg Am 2005;87: [28] Pitcher GM, Ritchie J, Henry JL. Nerve constriction in the rat: model of neuropathic, surgical and central pain. Pain 1999;83: [29] Pogatzki EM, Gebhart GF, Brennan TJ. Characterization of Adelta- and C-fibers innervating the plantar rat hind paw one day after an incision. J Neurophysiol 2002;87: [30] Pogatzki EM, Vandermeulen EP, Brennan TJ. Effect of plantar local anesthetic injection on dorsal horn neuron activity and pain behaviors caused by incision. Pain 2002;97: [31] Popping DM, Zahn PK, Van Aken HK, Dasch B, Boche R, Pogatzki-Zahn EM. Effectiveness and safety of postoperative pain management: a survey of 18, 925 consecutive patients between 1998 and 2006: a database analysis of prospectively raised data. Br J Anaesth 2008;101: [32] Quessy SN. The challenges of translational research for analgesics: the state of knowledge needs upgrading and some uncomfortable deficiencies remain to be urgently addressed. J Pain 2010;11:

37 S40 T.J. Brennan / PAIN Ò 152 (2011) S33 S40 [33] Reeh PW, Bayer J, Kocher L, Handwerker HO. Sensitization of nociceptive cutaneous nerve fibers from the rat s tail by noxious mechanical stimulation. Exp Brain Res 1987;65: [34] Rice AS, Cimino-Brown D, Eisenach JC, Kontinen VK, Lacroix-Fralish ML, Machin I, Mogil JS, Stohr T. Animal models and the prediction of efficacy in clinical trials of analgesic drugs: a critical appraisal and call for uniform reporting standards. Pain 2008;139: [35] Schmidt R, Schmelz M, Forster C, Ringkamp M, Torebjork E, Handwerker H. Novel classes of responsive and unresponsive C nociceptors in human skin. J Neurosci 1995;15: [36] Singelyn FJ, Deyaert M, Joris D, Pendeville E, Gouverneur JM. Effects of intravenous patient-controlled analgesia with morphine, continuous epidural analgesia, and continuous three-in-one block on postoperative pain and knee rehabilitation after unilateral total knee arthroplasty. Anesth Analg 1998;87: [37] Stubhaug A, Breivik H, Eide PK, Kreunen M, Foss A. Mapping of punctuate hyperalgesia around a surgical incision demonstrates that ketamine is a powerful suppressor of central sensitization to pain following surgery. Acta Anaesthesiol Scand 1997;41: [38] Vandermeulen EP, Brennan TJ. Alterations in ascending dorsal horn neurons by a surgical incision in the rat foot. Anesthesiology 2000;93: [39] Weber J, Loram L, Mitchell B, Themistocleous A. A model of incisional pain: the effects of dermal tail incision on pain behaviours of Sprague Dawley rats. J Neurosci Methods 2005;145: [40] Whiteside GT, Harrison J, Boulet J, Mark L, Pearson M, Gottshall S, Walker K. Pharmacological characterisation of a rat model of incisional pain. Br J Pharmacol 2004;141: [41] Xu J, Brennan TJ. Comparison of skin incision vs. skin plus deep tissue incision on ongoing pain and spontaneous activity in dorsal horn neurons. Pain 2009;144: [42] Xu J, Brennan TJ. Guarding pain and spontaneous activity of nociceptors after skin versus skin plus deep tissue incision. Anesthesiology 2010;112: [43] Xu J, Richebe P, Brennan TJ. Separate groups of dorsal horn neurons transmit spontaneous activity and mechanosensitivity one day after plantar incision. Eur J Pain 2009;13: [44] Zahn PK, Brennan TJ. Lack of effect of intrathecally administered N-methyl-Daspartate receptor antagonists in a rat model for postoperative pain. Anesthesiology 1998;88: [45] Zahn PK, Brennan TJ. Primary and secondary hyperalgesia in a rat model for human postoperative pain. Anesthesiology 1999;90: [46] Zahn PK, Pogatzki-Zahn EM, Brennan TJ. Spinal administration of MK-801 and NBQX demonstrates NMDA-independent dorsal horn sensitization in incisional pain. Pain 2005;114: [47] Zahn PK, Subieta A, Park SS, Brennan TJ. Effect of blockade of nerve growth factor and tumor necrosis factor on pain behaviors after plantar incision. J Pain 2004;5:

38 Promoting the knowledge of pain Become a Member What are the benefits of membership to you? Indirect affiliation to the International Association for the Study of Paid (IASP) without the expense of yearly international subscriptions Receive your quarterly Journal of Painsa Painsa Congresses (at reduced rates to Members) Regional Group symposia Contact through forums with other professionals in pain management Earn CPD points Is this your first application for membership? Yes No Are you a registered Health Professional? Yes No If yes, what is the nature of your practice? Professions Registration Number (if any) E.g. HPCSA. Title e.g. Prof / Dr / Mr / Mrs / Miss Surname First Name Address City Province Country Phone Land: Cell: Telefax Contact details: Phone: / Fax: / painsa@uiplay.com Annual Membership Fee: R Bona Fide Students: R only - proof required from Head of Department A direct deposit can be made into the account of: Painsa, Standard Bank,Northcliff, Account No , Branch Code Or send your cheque to: Painsa, P O Box 1105, Cramerview,

39

40

San Francisco Chronicle, June 2001

San Francisco Chronicle, June 2001 PAIN San Francisco Chronicle, June 2001 CONGENITAL INSENSITIVITY TO PAIN PAIN IS A SUBJECTIVE EXPERIENCE: It is not a stimulus MAJOR FEATURES OF THE PAIN EXPERIENCE: Sensory discriminative Affective (emotional)

More information

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

Spinal Cord Injury Pain. Michael Massey, DO CentraCare Health St Cloud, MN 11/07/2018 Spinal Cord Injury Pain Michael Massey, DO CentraCare Health St Cloud, MN 11/07/2018 Objectives At the conclusion of this session, participants should be able to: 1. Understand the difference between nociceptive

More information

Mechanism of Action & Physiologic Evidence

Mechanism of Action & Physiologic Evidence Mechanism of Action & Physiologic Evidence Treating Pain by Mitigating Central Sensitization BioElectronics Corporation Page 1 of 86 Background: The ActiPatch is a pulsed shortwave therapy (PSWT) product.

More information

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

Pain Pathways. Dr Sameer Gupta Consultant in Anaesthesia and Pain Management, NGH Pain Pathways Dr Sameer Gupta Consultant in Anaesthesia and Pain Management, NGH Objective To give you a simplistic and basic concepts of pain pathways to help understand the complex issue of pain Pain

More information

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

PAIN IS A SUBJECTIVE EXPERIENCE: It is not a stimulus. MAJOR FEATURES OF THE PAIN EXPERIENCE: Sensory discriminative Affective (emotional) Cognitive PAIN PAIN IS A SUBJECTIVE EXPERIENCE: It is not a stimulus MAJOR FEATURES OF THE PAIN EXPERIENCE: Sensory discriminative Affective (emotional) Cognitive MEASUREMENT OF PAIN: A BIG PROBLEM Worst pain ever

More information

Sensory Assessment of Regional Analgesia in Humans

Sensory Assessment of Regional Analgesia in Humans REVIEW ARTICLE Dennis M. Fisher, M.D., Editor-in-Chief Anesthesiology 2000; 93:1517 30 2000 American Society of Anesthesiologists, Inc. Lippincott Williams & Wilkins, Inc. Sensory Assessment of Regional

More information

PAIN MANAGEMENT in the CANINE PATIENT

PAIN MANAGEMENT in the CANINE PATIENT PAIN MANAGEMENT in the CANINE PATIENT Laurie Edge-Hughes, BScPT, MAnimSt (Animal Physio), CAFCI, CCRT Part 1: Laurie Edge-Hughes, BScPT, MAnimSt (Animal Physio), CAFCI, CCRT 1 Pain is the most common reason

More information

Sensory coding and somatosensory system

Sensory coding and somatosensory system Sensory coding and somatosensory system Sensation and perception Perception is the internal construction of sensation. Perception depends on the individual experience. Three common steps in all senses

More information

Pain. Pain. Pain: One definition. Pain: One definition. Pain: One definition. Pain: One definition. Psyc 2906: Sensation--Introduction 9/27/2006

Pain. Pain. Pain: One definition. Pain: One definition. Pain: One definition. Pain: One definition. Psyc 2906: Sensation--Introduction 9/27/2006 Pain Pain Pain: One Definition Classic Paths A new Theory Pain and Drugs According to the international Association for the Study (Merskey & Bogduk, 1994), Pain is an unpleasant sensory and emotional experience

More information

ANAT2010. Concepts of Neuroanatomy (II) S2 2018

ANAT2010. Concepts of Neuroanatomy (II) S2 2018 ANAT2010 Concepts of Neuroanatomy (II) S2 2018 Table of Contents Lecture 13: Pain and perception... 3 Lecture 14: Sensory systems and visual pathways... 11 Lecture 15: Techniques in Neuroanatomy I in vivo

More information

Prof Wayne Derman MBChB,BSc (Med)(Hons) PhD, FFIMS. Pain Management in the Elite Athlete: The 2017 IOC Consensus Statement

Prof Wayne Derman MBChB,BSc (Med)(Hons) PhD, FFIMS. Pain Management in the Elite Athlete: The 2017 IOC Consensus Statement Prof Wayne Derman MBChB,BSc (Med)(Hons) PhD, FFIMS Pain Management in the Elite Athlete: The 2017 IOC Consensus Statement 2 as 20 Experts published and leaders in their respective field 12 month lead in

More information

CHAPTER 10 THE SOMATOSENSORY SYSTEM

CHAPTER 10 THE SOMATOSENSORY SYSTEM CHAPTER 10 THE SOMATOSENSORY SYSTEM 10.1. SOMATOSENSORY MODALITIES "Somatosensory" is really a catch-all term to designate senses other than vision, hearing, balance, taste and smell. Receptors that could

More information

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

Proceedings of the World Small Animal Veterinary Association Sydney, Australia 2007 Proceedings of the World Small Animal Sydney, Australia 2007 Hosted by: Next WSAVA Congress REDUCING THE PAIN FACTOR AN UPDATE ON PERI-OPERATIVE ANALGESIA Sandra Forysth, BVSc DipACVA Institute of Veterinary,

More information

ANAT2010. Concepts of Neuroanatomy (II) S2 2018

ANAT2010. Concepts of Neuroanatomy (II) S2 2018 ANAT2010 Concepts of Neuroanatomy (II) S2 2018 Table of Contents Lecture 13: Pain and perception... 3 Lecture 14: Sensory systems and visual pathways... 11 Lecture 15: Techniques in Neuroanatomy I in vivo

More information

211MDS Pain theories

211MDS Pain theories 211MDS Pain theories Definition In 1986, the International Association for the Study of Pain (IASP) defined pain as a sensory and emotional experience associated with real or potential injuries, or described

More information

Virtually everyone has experienced pain in one

Virtually everyone has experienced pain in one Transfer of Advances in Sciences into Dental Education Recent Insights into Brainstem Mechanisms Underlying Craniofacial Pain Barry J. Sessle, B.D.S., M.D.S., B.Sc., Ph.D., F.R.S.C., D.Sc. (honorary) Abstract:

More information

Chapter 16. Sense of Pain

Chapter 16. Sense of Pain Chapter 16 Sense of Pain Pain Discomfort caused by tissue injury or noxious stimulation, and typically leading to evasive action important /// helps to protect us lost of pain in diabetes mellitus = diabetic

More information

By the end of this lecture the students will be able to:

By the end of this lecture the students will be able to: UNIT VII: PAIN Objectives: By the end of this lecture the students will be able to: Review the concept of somatosensory pathway. Describe the function of Nociceptors in response to pain information. Describe

More information

A Pain Clinic Approach to Fibromyalgia

A Pain Clinic Approach to Fibromyalgia A Pain Clinic Approach to Fibromyalgia Brigitte Gertoberens, Pain Medicine Specialist Malcolm Johnson,Clinical Psychologist Murray Hames, Senior Physiotherapist The Auckland Regional Pain Service Prevalence

More information

Nervous system Reflexes and Senses

Nervous system Reflexes and Senses Nervous system Reflexes and Senses Physiology Lab-4 Wrood Slaim, MSc Department of Pharmacology and Toxicology University of Al-Mustansyria 2017-2018 Nervous System The nervous system is the part of an

More information

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

Receptors and Neurotransmitters: It Sounds Greek to Me. Agenda. What We Know About Pain 9/7/2012 Receptors and Neurotransmitters: It Sounds Greek to Me Cathy Carlson, PhD, RN Northern Illinois University Agenda We will be going through this lecture on basic pain physiology using analogies, mnemonics,

More information

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

Somatosensory Physiology (Pain And Temperature) Richard M. Costanzo, Ph.D. Somatosensory Physiology (Pain And Temperature) Richard M. Costanzo, Ph.D. OBJECTIVES After studying the material of this lecture the student should be familiar with: 1. The relationship between nociception

More information

MYOFASCIAL PAIN. Dr. Janet Travell ( ) credited with bringing MTrPs to the attention of healthcare providers.

MYOFASCIAL PAIN. Dr. Janet Travell ( ) credited with bringing MTrPs to the attention of healthcare providers. Myofascial Trigger Points background info Laurie Edge-Hughes BScPT, MAnimSt (Animal Physio), CAFCI, CCRT History lesson Dr. Janet Travell (1901 1997) credited with bringing MTrPs to the attention of healthcare

More information

MANAGING CHRONIC PAIN

MANAGING CHRONIC PAIN George Hardas MANAGING CHRONIC PAIN The guide to understanding chronic pain and how to manage it. George Hardas MMed (UNSW) MScMed (Syd) MChiro (Macq) BSc (Syd) Grad Cert Pain Management (Syd) Cognitive

More information

Center for Sensory-Motor Interaction (SMI), Laboratory for Musculoskeletal Pain and Motor Control, Aalborg University, Denmark

Center for Sensory-Motor Interaction (SMI), Laboratory for Musculoskeletal Pain and Motor Control, Aalborg University, Denmark 14RC2 Assessment and mechanisms of musculo-skeletal pain Thomas Graven-Nielsen, Lars Arendt-Nielsen Center for Sensory-Motor Interaction (SMI), Laboratory for Musculoskeletal Pain and Motor Control, Aalborg

More information

Chapter 14: The Cutaneous Senses

Chapter 14: The Cutaneous Senses Chapter 14: The Cutaneous Senses Somatosensory System There are three parts Cutaneous senses - perception of touch and pain from stimulation of the skin Proprioception - ability to sense position of the

More information

Physiology of synapses and receptors

Physiology of synapses and receptors Physiology of synapses and receptors Dr Syed Shahid Habib Professor & Consultant Clinical Neurophysiology Dept. of Physiology College of Medicine & KKUH King Saud University REMEMBER These handouts will

More information

SOMATOSENSORY SYSTEMS AND PAIN

SOMATOSENSORY SYSTEMS AND PAIN SOMATOSENSORY SYSTEMS AND PAIN A 21 year old man presented with a stab wound of the right side of the neck (Panel A). Neurological examination revealed right hemiplegia and complete right-sided loss of

More information

Pain and Temperature Objectives

Pain and Temperature Objectives Pain and Temperature Objectives 1. Describe the types of sensory receptors that transmit pain and temperature. 2. Understand how axon diameter relates to transmission of pain and temp information. 3. Describe

More information

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

Pathophysiology of Pain. Ramon Go MD Assistant Professor Anesthesiology and Pain medicine NYP-CUMC Pathophysiology of Pain Ramon Go MD Assistant Professor Anesthesiology and Pain medicine NYP-CUMC Learning Objectives Anatomic pathway of nociception Discuss the multiple target sites of pharmacological

More information

Corporate Medical Policy

Corporate Medical Policy Corporate Medical Policy Intravenous Anesthetics for the Treatment of Chronic Pain File Name: Origination: Last CAP Review: Next CAP Review: Last Review: intravenous_anesthetics_for_the_treatment_of_chronic_pain

More information

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

What is Pain? An unpleasant sensory and emotional experience associated with actual or potential tissue damage. Pain is always subjective Pain & Acupuncture What is Pain? An unpleasant sensory and emotional experience associated with actual or potential tissue damage. NOCICEPTION( the neural processes of encoding and processing noxious stimuli.)

More information

Pain teaching. Muhammad Laklouk

Pain teaching. Muhammad Laklouk Pain teaching Muhammad Laklouk Definition Pain An unpleasant sensory and emotional experience associated with actual or potential tissue damage or described in terms of such damage. Sensory (discriminatiory)

More information

Chapter 6. Gathering information; the sensory systems

Chapter 6. Gathering information; the sensory systems Chapter 6 Gathering information; the sensory systems Gathering information the sensory systems The parts of the nervous system that receive and process information are termed sensory systems. There are

More information

Department of Neurology/Division of Anatomical Sciences

Department of Neurology/Division of Anatomical Sciences Spinal Cord I Lecture Outline and Objectives CNS/Head and Neck Sequence TOPIC: FACULTY: THE SPINAL CORD AND SPINAL NERVES, Part I Department of Neurology/Division of Anatomical Sciences LECTURE: Monday,

More information

Basic Neuroscience. Sally Curtis

Basic Neuroscience. Sally Curtis The Physiology of Pain Basic Neuroscience Sally Curtis sac3@soton.ac.uk The behaviour of humans is a result of the actions of nerves. Nerves form the basis of Thoughts, sensations and actions both reflex

More information

Neural Integration I: Sensory Pathways and the Somatic Nervous System

Neural Integration I: Sensory Pathways and the Somatic Nervous System 15 Neural Integration I: Sensory Pathways and the Somatic Nervous System PowerPoint Lecture Presentations prepared by Jason LaPres Lone Star College North Harris An Introduction to Sensory Pathways and

More information

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

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance Vol. 19 No. 1(Suppl.) January 2000 Journal of Pain and Symptom Management S7 Proceedings Supplement NDMA-Receptor Antagonists: Evolving Role in Analgesia NMDA-Receptor Antagonists and Opioid Receptor Interactions

More information

Overview of Questions

Overview of Questions Overview of Questions What are the sensors in the skin, what do they respond to and how is this transmitted to the brain? How does the brain represent touch information? What is the system for sensing

More information

Skin types: hairy and glabrous (e.g. back vs. palm of hand)

Skin types: hairy and glabrous (e.g. back vs. palm of hand) Lecture 19 revised 03/10 The Somatic Sensory System Skin- the largest sensory organ we have Also protects from evaporation, infection. Skin types: hairy and glabrous (e.g. back vs. palm of hand) 2 major

More information

Lecture 22: A little Neurobiology

Lecture 22: A little Neurobiology BIO 5099: Molecular Biology for Computer Scientists (et al) Lecture 22: A little Neurobiology http://compbio.uchsc.edu/hunter/bio5099 Larry.Hunter@uchsc.edu Nervous system development Part of the ectoderm

More information

Pain and endometriosis: How to optimize the medical management?

Pain and endometriosis: How to optimize the medical management? Pre-congress course Endometriosis sand pelvic pain 1st Society for endometriosis and uterine Disorders Meeting Paris, 7-9 May 2015 Pain and endometriosis: How to optimize the medical management? J. FAIDHERBE

More information

Management of Pain related to Spinal Cord Lesion

Management of Pain related to Spinal Cord Lesion Management of Pain related to Spinal Cord Lesion A Neurologist s Perspective Vincent Mok, MD Associate Professor Division of Neurology Department of Medicine and Therapeutics The Chinese University of

More information

SENSORY NERVOUS SYSTEM & SENSORY RECEPTORS. Dr. Ayisha Qureshi Professor MBBS, MPhil

SENSORY NERVOUS SYSTEM & SENSORY RECEPTORS. Dr. Ayisha Qureshi Professor MBBS, MPhil SENSORY NERVOUS SYSTEM & SENSORY RECEPTORS Dr. Ayisha Qureshi Professor MBBS, MPhil Sensory Deprivation Tank Is the world really as we perceive it? Is the world really as we perceive it? NO. The world

More information

SYLLABUS SPRING 2011 COURSE: NSC NEUROBIOLOGY OF PAIN

SYLLABUS SPRING 2011 COURSE: NSC NEUROBIOLOGY OF PAIN SYLLABUS NSC 4358 NEUROBIOLOGY OF PAIN SPRING 2011 1 SYLLABUS SPRING 2011 COURSE: NSC 4358 001 NEUROBIOLOGY OF PAIN Instructor: Aage R. Møller PhD E-mail: AMOLLER@UTDALLAS.EDU Class schedule: Main Campus:

More information

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

Chronic pain: We should not underestimate the contribution of neural plasticity. *Gwyn N Lewis 1, David A Rice 1,2 Chronic pain: We should not underestimate the contribution of neural plasticity *Gwyn N Lewis 1, David A Rice 1,2 1 Health and Rehabilitation Research Institute, AUT University, Auckland, New Zealand 2

More information

Pharmacology of Pain Transmission and Modulation

Pharmacology of Pain Transmission and Modulation Pharmacology of Pain Transmission and Modulation 2 Jürg Schliessbach and Konrad Maurer Nociceptive Nerve Fibers Pain is transmitted to the central nervous system via thinly myelinated Aδ and unmyelinated

More information

Mechanism of Pain Production

Mechanism of Pain Production Mechanism of Pain Production Pain conducting nerve fibers are small myelinated (A-delta) or unmyelinated nerve fibers (C-fibers). Cell bodies are in the dorsal root ganglia (DRG) or sensory ganglia of

More information

Chapter 11: Functional Organization of Nervous Tissue

Chapter 11: Functional Organization of Nervous Tissue Chapter 11: Functional Organization of Nervous Tissue I. Functions of the Nervous System A. List and describe the five major nervous system functions: 1. 2. 3. 4. 5. II. Divisions of the Nervous System

More information

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

The biochemical origin of pain: The origin of all pain is inflammation and the inflammatory response: Inflammatory profile of pain syndromes The biochemical origin of pain: The origin of all pain is inflammation and the inflammatory response: Inflammatory profile of pain syndromes 1 Medical Hypothesis 2007, Vol. 69, pp. 1169 1178 Sota Omoigui

More information

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

Seizure: the clinical manifestation of an abnormal and excessive excitation and synchronization of a population of cortical Are There Sharing Mechanisms of Epilepsy, Migraine and Neuropathic Pain? Chin-Wei Huang, MD, PhD Department of Neurology, NCKUH Basic mechanisms underlying seizures and epilepsy Seizure: the clinical manifestation

More information

How strong is it? What is it? Where is it? What must sensory systems encode? 9/8/2010. Spatial Coding: Receptive Fields and Tactile Discrimination

How strong is it? What is it? Where is it? What must sensory systems encode? 9/8/2010. Spatial Coding: Receptive Fields and Tactile Discrimination Spatial Coding: Receptive Fields and Tactile Discrimination What must sensory systems encode? How strong is it? What is it? Where is it? When the brain wants to keep certain types of information distinct,

More information

Spatial Coding: Receptive Fields and Tactile Discrimination

Spatial Coding: Receptive Fields and Tactile Discrimination Spatial Coding: Receptive Fields and Tactile Discrimination What must sensory systems encode? How strong is it? What is it? Where is it? When the brain wants to keep certain types of information distinct,

More information

Medical Neuroscience Tutorial

Medical Neuroscience Tutorial Pain Pathways Medical Neuroscience Tutorial Pain Pathways MAP TO NEUROSCIENCE CORE CONCEPTS 1 NCC1. The brain is the body's most complex organ. NCC3. Genetically determined circuits are the foundation

More information

Is mindfulness meditation an appropriate therapy in patients presenting with chronic pain?

Is mindfulness meditation an appropriate therapy in patients presenting with chronic pain? Is mindfulness meditation an appropriate therapy in patients presenting with chronic pain? Background Chronic pain affects between one-third and one-half of the UK adult 10.4% to 14.3% of cases being moderate-severely

More information

SOMATIC SENSATION PART I: ALS ANTEROLATERAL SYSTEM (or SPINOTHALAMIC SYSTEM) FOR PAIN AND TEMPERATURE

SOMATIC SENSATION PART I: ALS ANTEROLATERAL SYSTEM (or SPINOTHALAMIC SYSTEM) FOR PAIN AND TEMPERATURE Dental Neuroanatomy Thursday, February 3, 2011 Suzanne S. Stensaas, PhD SOMATIC SENSATION PART I: ALS ANTEROLATERAL SYSTEM (or SPINOTHALAMIC SYSTEM) FOR PAIN AND TEMPERATURE Reading: Waxman 26 th ed, :

More information

Neuropathic Pain in Palliative Care

Neuropathic Pain in Palliative Care Neuropathic Pain in Palliative Care Neuropathic Pain in Advanced Cancer Affects 40% of patients Multiple concurrent pains are common Often complex pathophysiology with mixed components Nocioceptive Neuropathic

More information

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

Pain. Types of Pain. Types of Pain 8/21/2013 Pain 1 Types of Pain Acute Pain Complex combination of sensory, perceptual, & emotional experiences as a result of a noxious stimulus Mediated by rapidly conducting nerve pathways & associated with increased

More information

Reasons for thinking so, according to von Frey:

Reasons for thinking so, according to von Frey: Reasons for thinking so, according to von Frey: The experiment consisted of sticking a pig bristle against the eye. Even with fairly light contact, the bristle was painful...... and a warm and cold bristle

More information

Chapter 9. Nervous System

Chapter 9. Nervous System Chapter 9 Nervous System Central Nervous System (CNS) vs. Peripheral Nervous System(PNS) CNS Brain Spinal cord PNS Peripheral nerves connecting CNS to the body Cranial nerves Spinal nerves Neurons transmit

More information

-12. -Osama Asa ad Khader. -Ameera Almomani. - Loay Zgoul

-12. -Osama Asa ad Khader. -Ameera Almomani. - Loay Zgoul -12 -Osama Asa ad Khader -Ameera Almomani - Loay Zgoul In this lecture we are going to talk about dermatomes, visceral sensation and pain disorders. Sorry for any mistakes though I tried my best to include

More information

Somatic Sensory System I. Background

Somatic Sensory System I. Background Somatic Sensory System I. Background A. Differences between somatic senses and other senses 1. Receptors are distributed throughout the body as opposed to being concentrated at small, specialized locations

More information

Memory Systems II How Stored: Engram and LTP. Reading: BCP Chapter 25

Memory Systems II How Stored: Engram and LTP. Reading: BCP Chapter 25 Memory Systems II How Stored: Engram and LTP Reading: BCP Chapter 25 Memory Systems Learning is the acquisition of new knowledge or skills. Memory is the retention of learned information. Many different

More information

ANATOMY AND PHYSIOLOGY OF NEURONS. AP Biology Chapter 48

ANATOMY AND PHYSIOLOGY OF NEURONS. AP Biology Chapter 48 ANATOMY AND PHYSIOLOGY OF NEURONS AP Biology Chapter 48 Objectives Describe the different types of neurons Describe the structure and function of dendrites, axons, a synapse, types of ion channels, and

More information

Chapter 13. The Nature of Muscle Spindles, Somatic Reflexes, and Posture

Chapter 13. The Nature of Muscle Spindles, Somatic Reflexes, and Posture Chapter 13 The Nature of Muscle Spindles, Somatic Reflexes, and Posture Nature of Reflexes A reflex is an involuntary responses initiated by a sensory input resulting in a change in the effecter tissue

More information

GENERAL PAIN DEFINITIONS

GENERAL PAIN DEFINITIONS I. OVERVIEW GENERAL PAIN DEFINITIONS Charles E. Argoff, MD CHAPTER 1 1. What is pain? Some dictionaries define pain as An unpleasant sensation, occurring in varying degrees of severity as a consequence

More information

Pathophysiological Classification of Pain

Pathophysiological Classification of Pain PATHOPHYSIOLOGY Overview Pathophysiological Classification of Pain Central sensitization/ dysfunctional pain Nociceptive pain - Somatic - Visceral Multiple pain mechanisms may coexist (mixed pain) Neuropathic

More information

Ch. 47 Somatic Sensations: Tactile and Position Senses (Reading Homework) - Somatic senses: three types (1) Mechanoreceptive somatic senses: tactile

Ch. 47 Somatic Sensations: Tactile and Position Senses (Reading Homework) - Somatic senses: three types (1) Mechanoreceptive somatic senses: tactile Ch. 47 Somatic Sensations: Tactile and Position Senses (Reading Homework) - Somatic senses: three types (1) Mechanoreceptive somatic senses: tactile and position sensations (2) Thermoreceptive senses:

More information

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

Pre-op Interventions to Mitigate Post-op Acute and Chronic Pain Pre-op Interventions to Mitigate Post-op Acute and Chronic Pain H A R S H A S H A N T H A N N A. M D, M S C A S S O C I A T E P R O F E S S O R D E P A R T M E N T O F A N E S T H E S I A C H R O N I C

More information

PAIN MODULATION. numerical value. adjectives. DR SYED SHAHID HABIB Professor & Consultant Dept. of Physiology College of Medicine & KKUH

PAIN MODULATION. numerical value. adjectives. DR SYED SHAHID HABIB Professor & Consultant Dept. of Physiology College of Medicine & KKUH PAIN MODULATION numerical value adjectives DR SYED SHAHID HABIB Professor & Consultant Dept. of Physiology College of Medicine & KKUH OBJECTIVES At the end of this lecture you should be able to describe:

More information

Cortical Control of Movement

Cortical Control of Movement Strick Lecture 2 March 24, 2006 Page 1 Cortical Control of Movement Four parts of this lecture: I) Anatomical Framework, II) Physiological Framework, III) Primary Motor Cortex Function and IV) Premotor

More information

The Physiology of the Senses Chapter 8 - Muscle Sense

The Physiology of the Senses Chapter 8 - Muscle Sense The Physiology of the Senses Chapter 8 - Muscle Sense www.tutis.ca/senses/ Contents Objectives... 1 Introduction... 2 Muscle Spindles and Golgi Tendon Organs... 3 Gamma Drive... 5 Three Spinal Reflexes...

More information

Biomechanics of Pain: Dynamics of the Neuromatrix

Biomechanics of Pain: Dynamics of the Neuromatrix Biomechanics of Pain: Dynamics of the Neuromatrix Partap S. Khalsa, D.C., Ph.D. Department of Biomedical Engineering The Neuromatrix From: Melzack R (1999) Pain Suppl 6:S121-6. NIOSH STAR Symposium May

More information

Chapter 7. The Nervous System: Structure and Control of Movement

Chapter 7. The Nervous System: Structure and Control of Movement Chapter 7 The Nervous System: Structure and Control of Movement Objectives Discuss the general organization of the nervous system Describe the structure & function of a nerve Draw and label the pathways

More information

Neuropathic pain, pain matrix dysfunction, and pain syndromes

Neuropathic pain, pain matrix dysfunction, and pain syndromes Neuropathic pain, pain matrix dysfunction, and pain syndromes MSTN121 - Neurophysiology Session 3 Department of Myotherapy Session objectives Describe the mechanism of nociceptive chronic pain. Define

More information

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

Somatic Sensation (MCB160 Lecture by Mu-ming Poo, Friday March 9, 2007) Somatic Sensation (MCB160 Lecture by Mu-ming Poo, Friday March 9, 2007) Introduction Adrian s work on sensory coding Spinal cord and dorsal root ganglia Four somatic sense modalities Touch Mechanoreceptors

More information

Plasticity of Cerebral Cortex in Development

Plasticity of Cerebral Cortex in Development Plasticity of Cerebral Cortex in Development Jessica R. Newton and Mriganka Sur Department of Brain & Cognitive Sciences Picower Center for Learning & Memory Massachusetts Institute of Technology Cambridge,

More information

Introduction to Neurobiology

Introduction to Neurobiology Biology 240 General Zoology Introduction to Neurobiology Nervous System functions: communication of information via nerve signals integration and processing of information control of physiological and

More information

Chronic Cervical and Shoulder pain

Chronic Cervical and Shoulder pain Chronic Cervical and Shoulder pain Dr Arif Ghazi BSc MBBS FCARCSI FFPMCAI FIPP Consultant in Pain Medicine Clinical Lead for Pain Services, Whipps Cross University Hospital, Barts Health Honorary Consultant

More information

Special Issue on Pain and Itch

Special Issue on Pain and Itch Special Issue on Pain and Itch Title: Recent Progress in Understanding the Mechanisms of Pain and Itch Guest Editor of the Special Issue: Ru-Rong Ji, PhD Chronic pain is a major health problem world-wide.

More information

Chapter 11 Introduction to the Nervous System and Nervous Tissue Chapter Outline

Chapter 11 Introduction to the Nervous System and Nervous Tissue Chapter Outline Chapter 11 Introduction to the Nervous System and Nervous Tissue Chapter Outline Module 11.1 Overview of the Nervous System (Figures 11.1-11.3) A. The nervous system controls our perception and experience

More information

Journal of Orthopaedic & Sports Physical Therapy. January 2012; Volume 42; Number 1; pp. 5-18

Journal of Orthopaedic & Sports Physical Therapy. January 2012; Volume 42; Number 1; pp. 5-18 1 Upper Cervical and Upper Thoracic Thrust Manipulation Versus Nonthrust Mobilization in Patients With Mechanical Neck Pain: A Multicenter Randomized Clinical Trial Journal of Orthopaedic & Sports Physical

More information

Motor Control, Pain, Somatic Dysfunction, Core Stability. Richard G. Schuster, DO Shawn Kerger, DO, FAOASM 19 September 2016 OMED 2016 Anaheim, CA

Motor Control, Pain, Somatic Dysfunction, Core Stability. Richard G. Schuster, DO Shawn Kerger, DO, FAOASM 19 September 2016 OMED 2016 Anaheim, CA Motor Control, Pain, Somatic Dysfunction, Core Stability Richard G. Schuster, DO Shawn Kerger, DO, FAOASM 19 September 2016 OMED 2016 Anaheim, CA How do we move? Decision to move (Executive or Cognitive

More information

EE 791 Lecture 2 Jan 19, 2015

EE 791 Lecture 2 Jan 19, 2015 EE 791 Lecture 2 Jan 19, 2015 Action Potential Conduction And Neural Organization EE 791-Lecture 2 1 Core-conductor model: In the core-conductor model we approximate an axon or a segment of a dendrite

More information

Joint Session with ACOFP, AOASM and AAO: Motor Control, Pain, Somatic Dysfunction, Core Stability. Shawn R. Kerger, DO, FAOSM Richard G.

Joint Session with ACOFP, AOASM and AAO: Motor Control, Pain, Somatic Dysfunction, Core Stability. Shawn R. Kerger, DO, FAOSM Richard G. Joint Session with ACOFP, AOASM and AAO: Motor Control, Pain, Somatic Dysfunction, Core Stability Shawn R. Kerger, DO, FAOSM Richard G. Schuster, DO Motor Control, Pain, Somatic Dysfunction, Core Stability

More information

3/7/2018. IASP updated definition of pain. Nociceptive Pain. Transduction. (Nociceptors) Transmission. (Peripheral nerve) Modulation

3/7/2018. IASP updated definition of pain. Nociceptive Pain. Transduction. (Nociceptors) Transmission. (Peripheral nerve) Modulation IASP updated definition of pain The Pain of Trauma and The Trauma of Pain: The Opioid Crisis is Not What You Think. Bennet Davis, M.D. Many people report pain in the absence of tissue damage or any likely

More information

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

Nervous System. Master controlling and communicating system of the body. Secrete chemicals called neurotransmitters Nervous System Master controlling and communicating system of the body Interacts with the endocrine system to control and coordinate the body s responses to changes in its environment, as well as growth,

More information

Is OnabotulinumtoxinA Good for Other Head and Face Pain? Disclosures BoNT/A for non- CM Botulinum neurotoxin (BoNT) in clinical use for headache >20

Is OnabotulinumtoxinA Good for Other Head and Face Pain? Disclosures BoNT/A for non- CM Botulinum neurotoxin (BoNT) in clinical use for headache >20 1 2 3 4 5 6 Is OnabotulinumtoxinA Good for Other Head and Face Pain? Disclosures BoNT/A for non- CM Botulinum neurotoxin (BoNT) in clinical use for headache >20 years Efficacy of BoNT type A (onabotulinumtoxina,

More information

STRUCTURAL ORGANIZATION OF THE NERVOUS SYSTEM

STRUCTURAL ORGANIZATION OF THE NERVOUS SYSTEM STRUCTURAL ORGANIZATION OF THE NERVOUS SYSTEM STRUCTURAL ORGANIZATION OF THE BRAIN The central nervous system (CNS), consisting of the brain and spinal cord, receives input from sensory neurons and directs

More information

Warm-Up. Label the parts of the neuron below.

Warm-Up. Label the parts of the neuron below. Warm-Up Label the parts of the neuron below. A B C D E F G Warm-Up 1. One neuron transmits a nerve impulse at 40 m/s. Another conducts at the rate of 1 m/s. Which neuron has a myelinated axon? 2. List

More information

Neuroscience with Pharmacology 2 Functions and Mechanisms of Reflexes. Prof Richard Ribchester

Neuroscience with Pharmacology 2 Functions and Mechanisms of Reflexes. Prof Richard Ribchester Neuroscience with Pharmacology 2 Functions and Mechanisms of Reflexes Prof Richard Ribchester René Descartes Cogito, ergo sum The 21st century still holds many challenges to Neuroscience and Pharmacology

More information

συν together απτειν to clasp 2h Neuroscience with Pharmacology Functions and Mechanisms of Reflexes Cogito, ergo sum ( I think therefore I am ) Down

συν together απτειν to clasp 2h Neuroscience with Pharmacology Functions and Mechanisms of Reflexes Cogito, ergo sum ( I think therefore I am ) Down 2h Neuroscience with Pharmacology Functions and Mechanisms of Reflexes Neuroscience is studied at many different levels: from brain, to system, network, neurone, synapse, and molecule... Top Up Down René

More information

Introduction to Computational Neuroscience

Introduction to Computational Neuroscience Introduction to Computational Neuroscience Lecture 7: Network models Lesson Title 1 Introduction 2 Structure and Function of the NS 3 Windows to the Brain 4 Data analysis 5 Data analysis II 6 Single neuron

More information

Syllabus. Questions may appear on any of the topics below: I. Multidimensional Nature of Pain

Syllabus. Questions may appear on any of the topics below: I. Multidimensional Nature of Pain Questions may appear on any of the topics below: I. Multidimensional Nature of Pain Syllabus A. Epidemiology 1. Pain as a public health problem with social, ethical, legal and economic consequences 2.

More information

Sensory information processing, somato-sensory systems

Sensory information processing, somato-sensory systems mm? Sensory information processing, somato-sensory systems Recommended literature 1. Kandel ER, Schwartz JH, Jessel TM (2000) Principles of Neural Science, McGraw-Hill, Ch. xx. 2. Berne EM, Levy MN, Koeppen

More information

TRANSCUTANEOUS ELECTRICAL NERVE STIMULATION (TENS) Dr. Mohammed TA, Omar, PhD, PT Rehabilitation Science Department CAMS-KSU

TRANSCUTANEOUS ELECTRICAL NERVE STIMULATION (TENS) Dr. Mohammed TA, Omar, PhD, PT Rehabilitation Science Department CAMS-KSU TRANSCUTANEOUS ELECTRICAL NERVE STIMULATION (TENS) Dr. Mohammed TA, Omar, PhD, PT Rehabilitation Science Department CAMS-KSU momarar@ksu.edu.sa Definition of TENS and current specifications Modes of TENS

More information

The anatomy and physiology of pain

The anatomy and physiology of pain The anatomy and physiology of pain Charlotte E Steeds Abstract Pain is an unpleasant experience that results from both physical and psychological responses to injury. A complex set of pathways transmits

More information

Chapter 7. Objectives

Chapter 7. Objectives Chapter 7 The Nervous System: Structure and Control of Movement Objectives Discuss the general organization of the nervous system Describe the structure & function of a nerve Draw and label the pathways

More information

Neural Basis of Motor Control

Neural Basis of Motor Control Neural Basis of Motor Control Central Nervous System Skeletal muscles are controlled by the CNS which consists of the brain and spinal cord. Determines which muscles will contract When How fast To what

More information

Various Types of Pain Defined

Various Types of Pain Defined Various Types of Pain Defined Pain: The International Association for the Study of Pain describes pain as, An unpleasant sensory and emotional experience associated with actual or potential tissue damage,

More information