Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Pain ; 20(10): 1199-1208, 2019 10.
Article in English | MEDLINE | ID: mdl-31005594

ABSTRACT

Cutaneous mechanical hyperalgesia can be induced in healthy volunteers in early phase analgesic studies to model central sensitization, a key mechanism of persistent pain. However, such hyperalgesia is short-lived (a matter of hours), and is used only for assessing only single drug doses. In contrast, postsurgical peri-incisional hyperalgesia may be more persistent and hence be a more useful model for the assessment of the efficacy of new analgesics. We undertook quantitative sensory testing in 18 patients at peri-incisional and nonoperated sites before open inguinal hernia repair and up to the 24th postsurgical week. The spatial extent of punctate hyperalgesia and brush allodynia at the peri-incisional site were greatest at weeks 2 and 4, but had resolved by week 24. Heat allodynia, suggestive of local inflammation or peripheral sensitization, was not observed; instead, there were deficits in cold and heat sensory detection that persisted until week 24. The findings suggest that central sensitization contributes significantly to mechanical hyperalgesia at the peri-incisional site. The prolonged duration of hyperalgesia would be advantageous as a pain model, but there was considerable variability of mechanical hyperalgesia in the cohort; the challenges of recruitment may limit its use to small, early phase analgesic studies. PERSPECTIVE: Peri-incisional mechanical hyperalgesia persists for ≥4 weeks after open inguinal hernia repair and reflects central sensitization; this may have usefulness as a model of chronic pain to assess the potential of antineuropathic analgesics.


Subject(s)
Central Nervous System Sensitization/physiology , Hernia, Inguinal/surgery , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Pain, Postoperative/physiopathology , Adult , Aged , Follow-Up Studies , Humans , Male , Middle Aged , Neuralgia/diagnosis , Pain Measurement , Pain, Postoperative/diagnosis
2.
Pain ; 152(12): 2844-2851, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22019137

ABSTRACT

Cutaneous inflammation induced by ultraviolet (UV) irradiation in the UV-B range has received significant recent interest as a translational inflammatory pain model. Changes in thermal and mechanical sensitivities in the area of primary hyperalgesia are well documented in both the rat and human UV-B models, but the occurrence of secondary mechanical hyperalgesia is controversial. We investigated the occurrence of secondary mechanical hyperalgesia in the rat UV-B model. Additionally, we investigated whether secondary hyperalgesia was enhanced by heat rekindling of UV-B-irradiated skin as a new rat inflammatory model of sensitisation with an enhanced central contribution. UV-B irradiation (1000 mJ/cm(2)) induced significant secondary mechanical hyperalgesia and allodynia that peaked at 48 h. Heat rekindling (45 °C stimulus for 5 min) of UV-B-irradiated skin at 24h further enhanced and prolonged this secondary mechanical hyperalgesia and allodynia, with a peak at 72 h. Heat rekindling also induced a significant mechanical hyperalgesia and allodynia on the contralateral hind paw, further suggesting the contribution of central sensitisation. Our data provide strong evidence for a central contribution in both the rat UV-B pain model and an enhanced contribution in the new model combining UV-B irradiation with heat rekindling. We also elucidate potential differences in the methods used by ourselves and others to obtain mechanical withdrawal thresholds in rats, which may explain the lack of secondary hyperalgesia in the rat UV-B model.


Subject(s)
Disease Models, Animal , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Inflammation Mediators/adverse effects , Ultraviolet Rays/adverse effects , Animals , Dermatitis/etiology , Dermatitis/physiopathology , Hyperalgesia/drug therapy , Inflammation Mediators/physiology , Male , Rats , Rats, Wistar
3.
Eur J Pain ; 15(10): 1040-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21576029

ABSTRACT

Current treatments of neuropathic pain arising from conditions such as nerve injury/compression are only partially effective, and limited in their use by side-effects. p38 mitogen-activated protein kinase (MAPK) is involved in the regulation and synthesis of inflammatory mediators, and is the target for a novel class of cytokine-suppressive anti-inflammatory drugs. p38 inhibitors may reduce neuronal sensitisation in preclinical models of neuropathic pain, particularly where there is a substantial inflammatory component. An exploratory, multicentre, double-blind, placebo-controlled, two-period, cross-over trial was undertaken to evaluate the effect of dilmapimod (SB-681323), a selective p38 MAPK inhibitor, on neuropathic pain symptoms and signs. Fifty patients with nerve trauma, radiculopathy or carpal tunnel syndrome were randomised; 43 patients completed the study. Eligible patients received oral dilmapimod and placebo twice daily for 2 weeks, with an intervening washout period of 2-4 weeks. Subjects attended weekly for efficacy and safety assessments, which included evaluation of daily and current pain intensity using an 11-point numerical rating scale (NRS), quantitative sensory testing, allodynia and global impression of change. There was a statistically significant reduction in the primary endpoint of average daily pain score during the second week of treatment among patients treated with dilmapimod (15 mg/day) compared to placebo using NRS [0.80; 95% CI (0.28, 1.33); p=0.0034]. A similar trend for effect was seen in some secondary endpoints. Dilmapimod was well tolerated, with no clinically relevant safety findings. p38 MAPK inhibitors merit further evaluation for neuropathic pain in larger clinical trials, particularly for clinically meaningful analgesic effect size.


Subject(s)
Analgesics/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Neuralgia/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidines/administration & dosage , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adult , Aged , Analgesics/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Female , Humans , Male , Middle Aged , Neuralgia/enzymology , Protein Kinase Inhibitors/adverse effects , Pyridones/adverse effects , Pyrimidines/adverse effects , Young Adult
4.
Cephalalgia ; 30(10): 1241-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20855370

ABSTRACT

Central sensitisation is a key mechanism of migraine; understanding its modulation by anti-migraine drugs is essential for rationalising treatment. We used an animal model of central trigeminal sensitisation to investigate neuronal responses to dural electrical stimulation as a putative electrophysiological marker of sensitisation and its modulation by ketorolac. In anaesthetised rats, responses of single convergent wide-dynamic range neurons of the spinal trigeminal nucleus to dural electrical simulation were recorded in parallel to their ongoing activity and responses to facial mechanical stimulation before and after a short-term dural application of an IS. Both ongoing activity and responses to dural electrical stimuli were enhanced by the inflammatory challenge, whereas neuronal thresholds to mechanical skin stimulation were reduced (p < .05, N = 12). Intravenous ketorolac (2 mg/kg, N = 6) reduced ongoing activity and responses to dural electrical stimulation, and increased mechanical thresholds versus vehicle controls (p < .05, N = 6). We conclude that neuronal responses to dural electrical stimulation can serve as a suitable marker which together with admitted electrophysiological signs can objectively detect central trigeminal sensitisation and its modulation by anti-migraine treatments in this preclinical model of migraine.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Ketorolac/pharmacology , Neurons/physiology , Trigeminal Nucleus, Spinal/physiology , Animals , Biomarkers/analysis , Disease Models, Animal , Dura Mater/drug effects , Dura Mater/physiology , Electric Stimulation , Inflammation/physiopathology , Male , Migraine Disorders/physiopathology , Neurons/drug effects , Rats , Rats, Wistar , Trigeminal Nucleus, Spinal/drug effects
5.
Neurotherapeutics ; 6(4): 749-54, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19789077

ABSTRACT

The translation of analgesic efficacy seen in preclinical pain models into the clinic is problematic and is associated with a number of factors that may result in the failure of clinical trials to detect the effect of investigational therapeutic agents. The use of translational pain biomarkers in phase I trials can potentially reduce some of these risks by measuring the interaction between the drug and its target (termed target engagement) in humans. To serve this purpose, sensory tests and other measures of pharmacological activity in nociceptive pathways need to be identified, based on the preclinical profile of the drug being tested and the feasibility of human assessments. Here we discuss some examples to assess the utility of sensory and related pain biomarkers in the early phase of evaluation of novel analgesics for confirmation of target engagement in humans. The emphasis is on the TRPV1 antagonists, but some other target mechanisms are also discussed in examining the validity of this approach.


Subject(s)
Analgesics/therapeutic use , Clinical Trials as Topic , Pain Measurement/methods , Sensation , Animals , Humans , Pain/drug therapy , TRPV Cation Channels/antagonists & inhibitors
6.
Clin J Pain ; 25(6): 469-76, 2009.
Article in English | MEDLINE | ID: mdl-19542793

ABSTRACT

OBJECTIVES: One limitation of neuropathic pain clinical trials is the often large and variable extent of response in the placebo group, possibly obscuring true medication effects. We pooled data from 252 individuals in the placebo arms of 3 clinical trials of lamotrigine in patients with neuropathic pain to examine the relationship of baseline patient and study site characteristics with 12-week change in the Pain Intensity Numerical Rating Scale score (DeltaPI-NRS). The 574 patients in the pooled lamotrigine treatment arms were used as a replication dataset. MATERIALS AND METHODS: We performed univariable and multivariable regression analysis of predictors of DeltaPI-NRS. Clinical factors examined were baseline pain intensity score (mean daily PI-NRS over the week prior to randomization), age, sex, diagnosis, prior and concurrent gabapentin use, prior and concurrent tricyclic antidepressant use, pain duration, variability of daily pain scores during the baseline week, and slope of daily pain scores over the baseline week. Site factors evaluated were study site, US geographic region, recruitment rate, and recruitment period. RESULTS: Baseline PI-NRS and site recruitment rate were independent predictors of the 12-week DeltaPI-NRS in the last observation carried forward, observed case, and repeated measures analyses. Patients with higher baseline PI-NRS scores had a significantly greater 12-week reduction in pain intensity than patients with lower baseline scores. Patients within sites with a faster recruitment rate also had a significantly greater reduction of pain intensity than those in sites with slower recruitment. DISCUSSION: These results suggest that both patient and study site characteristics can influence the response in the placebo arms of neuropathic pain studies.


Subject(s)
Anticonvulsants/therapeutic use , Neuralgia/drug therapy , Placebo Effect , Triazines/therapeutic use , Aged , Dose-Response Relationship, Drug , Female , Humans , Lamotrigine , Male , Middle Aged , Models, Statistical , Neuralgia/psychology , Pain Clinics , Predictive Value of Tests , Randomized Controlled Trials as Topic , Regression Analysis , Time Factors
7.
Eur J Pain ; 13(9): 942-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19117776

ABSTRACT

Allodynia is a common, troublesome feature of neuropathic pain conditions. In a previous study of postherpetic neuralgia we observed that repeated tactile stimulation appeared to reduce the size of the area of allodynia in some patients. We have undertaken a pragmatic clinical study to characterise this phenomenon in neuropathic pain patients with a range of different aetiologies. Neuropathic pain patients with a discrete area of tactile allodynia were recruited (n=20). We assessed the sensitive area using punctate and dynamic tactile stimuli, and thermal quantitative sensory testing. On two separate testing visits, the patients had repeated (10x over 1 min) noxious heat or cotton bud strokes applied to the affected site or contralaterally. Tactile stimulation of the affected area evoked pain (median 7 NRS) and a reduction (>30%) in the area of allodynia in 9/18 patients (maximum -48+/-9%, after 20 min), although the intensity of allodynic pain was unchanged. This effect persisted for over 1h and was present the following day in all patients tested (n=5/5). No subjects showed an increase in area after allodynic stimulation. There was no change in heat pain threshold at a distant site following allodynic stimulation, suggesting no activation of diffuse noxious inhibitory control. Repeated thermal noxious stimulation (median NRS 7) could also elicit changes (>30%) in the area of allodynia in some patients (reductions in 7/20, increases in 3/20). Thus, we have found that a brief period of homotopic painful stimulation can reduce the area of allodynia in around half of patients with established neuropathic pains.


Subject(s)
Hyperalgesia/therapy , Nociceptors/physiology , Peripheral Nervous System Diseases/therapy , Physical Therapy Modalities , Touch/physiology , Adult , Aged , Aged, 80 and over , Female , Humans , Hyperalgesia/physiopathology , Male , Middle Aged , Neural Inhibition/physiology , Pain Measurement , Pain Threshold/physiology , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Physical Stimulation , Skin/innervation , Skin/physiopathology , Treatment Outcome
8.
Brain Res Rev ; 60(1): 243-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19168094

ABSTRACT

The pharmaceutical industry faces tough times. Despite tremendous advances in the science and technology of new lead identification and optimization, attrition rates for novel drug candidates making it into the clinic remain unacceptably high. A seamless boundary between basic preclinical and clinical arms of the discovery process, embodying the concept of 'translational research' is viewed by many as the way forward. The rational application of human experimental pain models in early clinical development is reviewed. Capsaicin, UV-irradiation and electrical stimulation methods have each been used to establish experimental hyperalgesia in Phase-I human volunteers and the application of these approaches is discussed in the context of several pharmacological examples. However, data generated from such studies must be integrated into a well-conceived and executed series of Phase-II efficacy trials in patients in order to derive maximal benefit. The challenges involved in optimal Phase-II/III trial design are reviewed with specific attention to the issues of sample size and placebo response. Finally, the application and potential of cortical EEG studies are discussed as an objective alternative to more conventional pain assessment tools with specific examples of how this technique has been applied to the study of NSAID and opiate-based therapeutics.


Subject(s)
Analgesics/pharmacology , Clinical Trials as Topic/standards , Clinical Trials as Topic/trends , Drug Design , Pain/drug therapy , Pain/physiopathology , Analgesics/chemical synthesis , Clinical Trials as Topic/methods , Humans , Models, Neurological , Neuropharmacology/methods , Neuropharmacology/standards , Neuropharmacology/trends , Outcome Assessment, Health Care/methods , Pain Measurement/methods , Patient Selection , Physical Stimulation/methods
9.
Drug Discov Today ; 14(1-2): 56-67, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19063991

ABSTRACT

TRPV1 is a noxious heat, capsaicin (vanilloid) and acid receptor for which the development of antagonists represents a novel therapeutic approach for the treatment of pain. TRPV1 antagonists have entered early clinical development and initial reports indicate that they have demonstrated pharmacodynamic effects consistent with TRPV1 antagonist activity and anti-hyperalgesic action in humans. Should these effects extend to the relief of symptoms experienced by patients with chronic pain then this class of compounds may offer one of the first novel mechanisms of action for the treatment for pain for many years. In this article we will discuss recent progress and challenges in the field in this highly competitive area of drug discovery.


Subject(s)
Analgesics/pharmacology , Pain/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Analgesics/therapeutic use , Animals , Chronic Disease , Drug Delivery Systems , Drug Design , Humans , Pain/physiopathology
11.
BMC Neurol ; 7: 21, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17683543

ABSTRACT

BACKGROUND: The Contact Heat Evoked Potential Stimulator (CHEPS) rapidly stimulates cutaneous small nerve fibres, and resulting evoked potentials can be recorded from the scalp. We have studied patients with symptoms of sensory neuropathy and controls using CHEPS, and validated the findings using other objective measures of small nerve fibres i.e. the histamine-induced skin flare response and intra-epidermal fibres (IEF), and also quantitative sensory testing (QST), a subjective measure. METHODS: In patients with symptoms of sensory neuropathy (n = 41) and healthy controls (n = 9) we performed clinical examination, QST (monofilament, vibration and thermal perception thresholds), nerve conduction studies, histamine-induced skin flares and CHEPS. Skin punch biopsies were immunostained using standard ABC immunoperoxidase for the nerve marker PGP 9.5 or the heat and capsaicin receptor TRPV1. Immunoreactive IEF were counted per length of tissue section and epidermal thickness recorded. RESULTS: Amplitudes of Adelta evoked potentials (muV) following face, arm or leg stimulation were reduced in patients (e.g. for the leg: mean +/- SEM - controls 11.7 +/- 1.95, patients 3.63 +/- 0.85, p = 0.0032). Patients showed reduced leg skin flare responses, which correlated with Adelta amplitudes (rs = 0.40, p = 0.010). In patient leg skin biopsies, PGP 9.5- and TRPV1-immunoreactive IEF were reduced and correlated with Adelta amplitudes (PGP 9.5, rs = 0.51, p = 0.0006; TRPV1, rs = 0.48, p = 0.0012). CONCLUSION: CHEPS appears a sensitive measure, with abnormalities observed in some symptomatic patients who did not have significant IEF loss and/or QST abnormalities. Some of the latter patients may have early small fibre dysfunction or ion channelopathy. CHEPS provides a clinically practical, non-invasive and objective measure, and can be a useful additional tool for the assessment of sensory small fibre neuropathy. Although further evaluation is required, the technique shows potential clinical utility to differentiate neuropathy from other chronic pain states, and provide a biomarker for analgesic development.


Subject(s)
Evoked Potentials, Somatosensory/physiology , Nerve Fibers/physiology , Peripheral Nervous System Diseases/pathology , Sensory Thresholds/physiology , Skin/innervation , Adult , Aged , Biopsy/methods , Electroencephalography/methods , Female , Histamine/pharmacology , Humans , Male , Middle Aged , Nerve Fibers/radiation effects , Peripheral Nervous System Diseases/physiopathology , Reaction Time/physiology , Sensory Thresholds/drug effects , Skin/physiopathology , Statistics, Nonparametric , TRPV Cation Channels/metabolism , Ubiquitin Thiolesterase/metabolism
12.
Pain ; 132(1-2): 132-41, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17659837

ABSTRACT

TRPV1 is a cation channel activated by a range of noxious stimuli and highly expressed in nociceptive fibres. TRPV1 receptors are involved in pain and sensitisation associated with tissue injury and inflammation; hence, TRPV1 antagonists are potentially useful for the treatment of such pain states. SB-705498 is a potent, selective and orally bioavailable TRPV1 antagonist with demonstrated efficacy in a number of preclinical pain models. In this first-time-into-human study, we have investigated the pharmacodynamic and antihyperalgesic activity of SB-705498. The compound was safe and well tolerated at single oral doses up to 400mg. In a cohort of 19 healthy volunteers, we used a randomised placebo-controlled single-blind cross-over design to assess the effects of SB-705498 (400mg) on heat-evoked pain and skin sensitisation induced by capsaicin or UVB irradiation. Compared with placebo, SB-705498 reduced the area of capsaicin-evoked flare (P=0.0047). The heat pain threshold on non-sensitised skin was elevated following SB-705498 (estimated difference from placebo [95% confidence intervals]: 1.3 degrees C [0.07,2.53], P=0.019). Following capsaicin sensitisation, the heat pain threshold and tolerance were similar between SB-705498 and placebo. However, SB-705498 increased heat pain tolerance at the site of UVB-evoked inflammation (estimated difference from placebo: 0.93 degrees C [0.25,1.6], P=0.0054). The magnitude of the pharmacodynamic effects of SB-705498 appeared to be related to plasma concentration. These results indicate that SB-705498, at a clinically safe and well-tolerated dose, has target-specific pharmacodynamic activity in humans. These data provide the first clinical evidence that a TRPV1 antagonist may alleviate pain and hyperalgesia associated with inflammation and tissue injury.


Subject(s)
Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Pain Measurement/drug effects , Pyrrolidines/administration & dosage , Signal Transduction/drug effects , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Administration, Oral , Adolescent , Adult , Analgesics/administration & dosage , Cohort Studies , Dose-Response Relationship, Drug , Female , Hot Temperature , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Male , Middle Aged , Placebo Effect , Treatment Outcome , Urea/administration & dosage
13.
Eur J Neurosci ; 25(12): 3576-82, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17610577

ABSTRACT

Fibromyalgia is characterized by chronic widespread pain and bodily tenderness and is often accompanied by affective disturbances. Accumulating evidence indicates that fibromyalgia may involve a dysfunction of modulatory systems in the brain. While brain dopamine is best known for its role in pleasure, motivation and motor control, recent evidence suggests that it is also involved in pain modulation. Because dopamine is implicated in both pain modulation and affective processing, we hypothesized that fibromyalgia may involve a disturbance of dopaminergic neurotransmission. Fibromyalgia patients and matched healthy control subjects were subjected to deep muscle pain produced by injection of hypertonic saline into the anterior tibialis muscle. In order to determine the endogenous release of dopamine in response to painful stimulation, we used positron emission tomography to examine binding of [(11)C]-raclopride (D2/D3 ligand) in the brain during injection of painful hypertonic saline and nonpainful normal saline. Fibromyalgia patients experienced the hypertonic saline as more painful than healthy control subjects. Control subjects released dopamine in the basal ganglia during the painful stimulation, whereas fibromyalgia patients did not. In control subjects, the amount of dopamine release correlated with the amount of perceived pain but in fibromyalgia patients no such correlation was observed. These findings provide the first direct evidence that fibromyalgia patients have an abnormal dopamine response to pain. The disrupted dopaminergic reactivity in fibromyalgia patients could be a critical factor underlying the widespread pain and discomfort in fibromyalgia and suggests that the therapeutic effects of dopaminergic treatments for this intractable disorder should be explored.


Subject(s)
Dopamine/metabolism , Fibromyalgia/metabolism , Fibromyalgia/psychology , Pain/metabolism , Adult , Analysis of Variance , Area Under Curve , Brain Mapping , Carbon Isotopes/pharmacokinetics , Case-Control Studies , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Dopamine Antagonists/pharmacokinetics , Female , Fibromyalgia/diagnostic imaging , Humans , Middle Aged , Pain/chemically induced , Pain/diagnostic imaging , Pain Measurement/methods , Positron-Emission Tomography/methods , Psychophysics , Raclopride/pharmacokinetics , Saline Solution, Hypertonic
14.
J Psychopharmacol ; 21(3): 259-71, 2007 May.
Article in English | MEDLINE | ID: mdl-17591654

ABSTRACT

Ketamine is a dissociative anaesthetic that has been used in the clinic for many years. At low, sub-anaesthetic doses, it is a relatively selective and potent antagonist of the N-methyl-D-aspartate (NMDA) receptor. It belongs to the class of uncompetitive antagonists and blocks the receptor by binding to a specific site within the NMDA receptor channel when it is open. Like other compounds of this class, ketamine can cause hallucinations or other untoward central effects which limit its use in the clinic. Nevertheless, because of the evidence on the importance of NMDA receptor-mediated pLasticity in chronic pain, low doses of ketamine have been explored in a wide range of pain conditions. The majority of studies with ketamine have shown efficacy; however, it has not been possible to separate safely the pain relief from the side effects of the drug. Hence, clinical use of ketamine as a pain treatment is very limited. Nevertheless, ketamine has served as a useful tool to provide a compelling rationale for developing other NMDA antagonists. Some of the new compounds of this class, particularly those acting at the NR2B subtype of the NMDA receptor, have shown promise in preclinical and clinical studies.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Ketamine/pharmacology , Ketamine/therapeutic use , Neuronal Plasticity/drug effects , Pain/drug therapy , Pain/physiopathology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/drug effects , Animals , Excitatory Amino Acid Antagonists/adverse effects , Humans , Ketamine/adverse effects
16.
J Neurosci ; 27(15): 4004-7, 2007 Apr 11.
Article in English | MEDLINE | ID: mdl-17428976

ABSTRACT

Fibromyalgia is an intractable widespread pain disorder that is most frequently diagnosed in women. It has traditionally been classified as either a musculoskeletal disease or a psychological disorder. Accumulating evidence now suggests that fibromyalgia may be associated with CNS dysfunction. In this study, we investigate anatomical changes in the brain associated with fibromyalgia. Using voxel-based morphometric analysis of magnetic resonance brain images, we examined the brains of 10 female fibromyalgia patients and 10 healthy controls. We found that fibromyalgia patients had significantly less total gray matter volume and showed a 3.3 times greater age-associated decrease in gray matter than healthy controls. The longer the individuals had had fibromyalgia, the greater the gray matter loss, with each year of fibromyalgia being equivalent to 9.5 times the loss in normal aging. In addition, fibromyalgia patients demonstrated significantly less gray matter density than healthy controls in several brain regions, including the cingulate, insular and medial frontal cortices, and parahippocampal gyri. The neuroanatomical changes that we see in fibromyalgia patients contribute additional evidence of CNS involvement in fibromyalgia. In particular, fibromyalgia appears to be associated with an acceleration of age-related changes in the very substance of the brain. Moreover, the regions in which we demonstrate objective changes may be functionally linked to core features of the disorder including affective disturbances and chronic widespread pain.


Subject(s)
Aging, Premature/pathology , Cerebral Cortex/pathology , Fibromyalgia/pathology , Adult , Brain/pathology , Brain Mapping/methods , Female , Humans , Middle Aged , Time Factors
17.
J Pain ; 8(5): 443-51, 2007 May.
Article in English | MEDLINE | ID: mdl-17337250

ABSTRACT

UNLABELLED: Inflammation and trauma lead to tissue acidification and release of inflammatory mediators, including prostaglandin E2 (PGE2). Protons can evoke pain through acid-sensing ion channels (ASICs) and TRPV1 receptors. In this study, we examined whether PGE2 can potentiate proton-induced nociception in humans on injection into skin and muscle. Psychophysical and vascular responses to microinjections of protons (pH 6.0 and 6.5), PGE2 (10-6 and 10-7 M) and their combinations into forearm skin (30 microL) or anterior tibial muscle (50 microL) were assessed in 16 male subjects. Pain intensity, axon reflex erythema, and heat pain thresholds were recorded after skin challenge; pain intensity and thresholds for pressure-evoked pain were recorded after intramuscular injections. Intradermal or intramuscular injections of PGE2 induced very low levels of pain similar to saline. Administration of low pH caused moderate pain within 5 seconds that declined rapidly over 15 to 20 seconds. In comparison, coinjection of low pH with PGE2 led to a biphasic profile of the pain response. Combined pH + PGE2 stimulation provoked significantly increased pain in the second phase after injections (20 to 100 seconds) both in skin and muscle, whereas the initial injection pain was not enhanced. Heat pain thresholds were reduced after PGE2 and combined pH + PGE2, whereas flare responses were rather attenuated on coadministration of low pH with PGE2. Intriguingly, when compared with skin, muscle pain was significantly lower in the initial phase (0 to 15 seconds) but significantly higher in the second phase (20 to 100 seconds after injection). PERSPECTIVE: PGE2 can potentiate nociceptor activation by protons in human skin and muscle, indicated by increased sustained pain ratings. This can be best explained by TRPV1 sensitization in the presence of PGE2, a mechanism potentially relevant for inflammatory and injury-induced pain.


Subject(s)
Dinoprostone/therapeutic use , Oxytocics/therapeutic use , Pain/drug therapy , Protons/adverse effects , Adult , Analysis of Variance , Dose-Response Relationship, Drug , Drug Interactions , Humans , Male , Muscle, Skeletal/innervation , Pain/chemically induced , Pain Measurement/methods , Pain Threshold/drug effects , Skin/innervation , Time Factors , Vasodilation/drug effects
18.
Neuropharmacology ; 51(1): 12-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16616769

ABSTRACT

Clinical reports have described a long-lasting relief in neuropathic pain patients treated with NMDA receptor antagonists; it is unclear, however, what mediates this effect. In this work, we have used two NMDA antagonists of different class to investigate if the antiallodynic effects in a rat neuropathy model can outlast their in vivo NMDA antagonism. Both the uncompetitive NMDA antagonist ketamine and the glycine(B) antagonist MRZ 2/576 inhibited neuronal responses to iontophoretic NMDA in anaesthetised rats with the time course consistent with their known pharmacokinetics (t(1/2) approximately 10-12min, similar in control and nerve-injured rats). Surprisingly, the antiallodynic effects of the same doses of the NMDA antagonists in the neuropathic pain model were long-lasting (>3h with ketamine, >24h with MRZ 2/576). The effect of ketamine was further prolonged (>24h) when combined with a short-acting opioid, fentanyl, which only produced a short effect ( approximately 40min) when given alone. The duration of centrally mediated side effects of ketamine and MRZ 2/576 was short, similar to the in vivo NMDA antagonism. We speculate that NMDA receptor blockade may down-regulate the central sensitisation triggered by nerve injury, resulting in a long-lasting antiallodynic effect. Development of short-acting NMDA antagonists could represent a strategy for improving their tolerability.


Subject(s)
Pain/drug therapy , Peripheral Nervous System Diseases/complications , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Cold Temperature , Constriction, Pathologic/complications , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Fentanyl/pharmacology , Iontophoresis , Ketamine/pharmacology , Male , Motor Activity/drug effects , N-Methylaspartate/administration & dosage , N-Methylaspartate/pharmacology , Pain/etiology , Phthalazines/pharmacology , Posterior Horn Cells/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
19.
Brain Res ; 1048(1-2): 218-27, 2005 Jun 28.
Article in English | MEDLINE | ID: mdl-15922311

ABSTRACT

NMDA receptors are implicated in central sensitisation underlying chronic pain, and NMDA antagonists have a potential for the treatment of neuropathic pain. Functional NMDA receptors are also present on primary afferents, where they may play a role in pro-nociceptive plasticity. The importance of this mechanism in neuropathic pain remains unclear. In the present work, we have compared in models of chronic pain the effects of NMDA antagonists at the glycine(B) site with different central access. L-701,324 (the centrally active antagonist) and 5,7-dichlorokynurenic acid (5,7-DCK, known to have limited central access) were tested after systemic administration in rats in the formalin test and in two models of neuropathic pain. The ability of these compounds to exert central actions (sedation, ataxia) was tested in the open field locomotion test; central NMDA antagonism in vivo was tested in anaesthetised rats on responses of spinal cord neurones to iontophoretic NMDA. Both L-701,324 (2.15-21.5 mg/kg i.p.) and 5,7-DCK (10-46.4 mg/kg i.v.) dose-dependently inhibited Phase II of formalin-evoked behaviour. Likewise, both compounds reversed cold allodynia in the chronic constriction injury model and tactile allodynia in animals with spinal nerve ligation. However, only L-701,324 was able to inhibit neuronal responses to NMDA in the antihyperalgesic dose range; 5,7-DCK was inactive on NMDA responses up to 46.4 mg/kg i.v. or 68.1 mg/kg i.p. Consistent with the lack of inhibition of central NMDA-evoked activity, 5,7-DCK did not alter spontaneous behaviour in the open field test, whereas it was significantly inhibited by L-701,324. Thus, peripheral NMDA receptors may substantially contribute to the efficacy of NMDA antagonists in neuropathic pain.


Subject(s)
Excitatory Amino Acid Antagonists/administration & dosage , Kynurenic Acid/analogs & derivatives , Neuralgia/drug therapy , Quinolones/administration & dosage , Action Potentials/drug effects , Animals , Constriction, Pathologic/complications , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Interactions , Excitatory Amino Acid Agonists/pharmacology , Kynurenic Acid/administration & dosage , Ligation/methods , Male , N-Methylaspartate/pharmacology , Neuralgia/etiology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Nerves/injuries , Spinal Nerves/physiopathology , Time Factors
20.
Anesthesiology ; 101(6): 1400-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564948

ABSTRACT

BACKGROUND: In chronic pain, increased activity from intact or damaged peripheral nerve endings results in an enhanced response in central pain transmission systems, a mechanism known as central sensitization. Central sensitization can also be invoked in human experimental models. Therefore, these models may be useful to characterize novel analgesics in humans. The anticonvulsant gabapentin has demonstrated efficacy in patients with neuropathic pain, but its mode of action remains unclear. This study examined the effects of gabapentin on signs of central sensitization (brush and pinprick hyperalgesia) in a human model of capsaicin-evoked pain, using a gabapentin dosing regimen similar to that used in the clinic. The aims were to determine whether gabapentin, dosed in a manner similar to that used in the clinic, affected the various components of central sensitization and to assess the utility of this model for characterizing novel analgesics. METHODS: Intradermal capsaicin (100 microg/20 microl) was administered in the volar forearm of 41 male human volunteers to induce pain and clinical signs of central sensitization. Gabapentin (titrated to 2,400 mg daily) or placebo was given orally for 15 days in a randomized, double-blind, parallel-group design. The capsaicin test was conducted at baseline and after gabapentin or placebo. Endpoints were the size of areas of brush-evoked allodynia (with cotton gauze) and pinprick hyperalgesia (with von Frey filament), and the intensity of ongoing brush- and pinprick-evoked pain. RESULTS: Gabapentin significantly reduced the area of brush allodynia compared with placebo (P

Subject(s)
Amines/therapeutic use , Analgesics/therapeutic use , Central Nervous System/drug effects , Cyclohexanecarboxylic Acids/therapeutic use , Hyperalgesia/drug therapy , gamma-Aminobutyric Acid/therapeutic use , Adult , Amines/adverse effects , Amines/blood , Analgesics/adverse effects , Analgesics/blood , Capsaicin , Cyclohexanecarboxylic Acids/adverse effects , Cyclohexanecarboxylic Acids/blood , Double-Blind Method , Gabapentin , Humans , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Injections, Intradermal , Male , Pain Measurement/drug effects , Physical Stimulation , gamma-Aminobutyric Acid/adverse effects , gamma-Aminobutyric Acid/blood
SELECTION OF CITATIONS
SEARCH DETAIL