Вы находитесь на странице: 1из 12

REVIEWS

Mechanisms, impact and management


of pain in rheumatoid arthritis
David A. Walsh and Daniel F. McWilliams
Abstract | People with rheumatoid arthritis (RA) identify pain as their most important symptom, one that
often persists despite optimal control of inflammatory disease. RA pain arises from multiple mechanisms,
involving inflammation, peripheral and central pain processing and, with disease progression, structural
change within the joint. Consequently, RA pain has a wide range of characteristics—constant or intermittent,
localized or widespread—and is often associated with psychological distress and fatigue. Dominant pain
mechanisms in an individual are identified by critical evaluation of clinical symptoms and signs, and by
laboratory and imaging tests. Understanding these mechanisms is essential for effective management,
although evidence from preclinical models should be interpreted with caution. A range of pharmacological
analgesic and immunomodulatory agents, psychological interventions and surgery may help manage RA pain.
Pain contributes importantly to the clinical assessment of inflammatory disease activity, and noninflammatory
components of RA pain should be considered when gauging eligibility for or response to biologic agents.
Further randomized controlled trials are required to determine the optimal usage of analgesics in RA, and novel
agents with greater efficacy and lower propensity for adverse events are urgently needed. Meanwhile, targeted
use of existing treatments could reduce pain in people with RA.
Walsh, D. A. & McWilliams, D. F. Nat. Rev. Rheumatol. 10, 581–592 (2014); published online 27 May 2014; doi:10.1038/nrrheum.2014.64

Introduction
Pain is multidimensional, defined by the International arthritis.5 Pain in patients with active inflammatory dis­
Association for the Study of Pain as “an unpleasant sensory ease is sometimes described as ‘aching’, ‘sharp’, ‘throb-
and emotional experience associated with actual or poten- bing’, ‘tender’ and ‘shooting’,5 or as ‘tiring’ and ‘sickening’,6
tial tissue damage, or described in terms of such damage.”1 whereas those with less-active inflammatory disease have
People with rheumatoid arthritis (RA) commonly high- highlighted words such as ‘gnawing’, ‘hurting’, ‘tender’
light pain as their most important problem.2 Pain can be and ‘dullness’.7
associated with psychological distress, can impair physi- People with RA report some pain qualities that are char-
cal and social functioning, and can increase health-care acteristic of pain arising from nervous system pathology
utilization.3 The prognosis of pain in RA is often poor, ­—neuropathic pain (see Box 1 for definitions of rele­vant
even when inflammatory disease is optimally controlled.4 pain terms).8 ‘Burning’, pain on light pressure or sudden
Better treatment of RA pain is urgently needed. pain attacks likened to electric shocks can be of suffi-
In this Review, we describe pain in RA and summarize cient severity in RA to permit classification by screen-
contemporary knowledge about mechanisms, associated ing questionnaires as neuropathic pain (36% in one 2013
factors and management strategies. We also highlight study 9). People with RA can experience true neuro­pathic
issues where further study could lead to improvements pain (for example, due to carpal tunnel syndrome),
in clinical care. but neuropathic-like symptoms might other­wise indicate
pain mechanisms common to both RA and neuropathic
Qualities of RA pain pain, as also found in fibromyalgia10 and osteoarthritis
Pain can be assessed and measured using a variety of (OA) without specific neuro­pathology.11 Neuropathic-
Arthritis Research UK validated questionnaires, some of which—those com- like symptoms might be associated with abnormal cen­
Pain Centre, Academic monly used in RA—are summarized in Table 1. The pain tral pain processing, indicated by widespread reductions
Rheumatology,
University of associated with RA is described using the full range of in pain thresholds in response to pressure and increased
Nottingham, Clinical terms encompassed in the McGill Pain Questionnaire5, cerebral activity in response to painful stimuli.10
Sciences Building,
City Hospital,
with ‘aching’, ‘sharp’, ‘throbbing’, ‘tender’ and ‘shooting’ RA pain has various temporal patterns, including
Hucknall Road, being used most frequently in one study of patients with occur­rences of pain restricted to weight-bearing or joint
Nottingham NG5 1PB, move­ment, episodic flares, or constant pain. People with
UK (D.A.W., D.F.M.).
RA clearly distinguish between constant and intermittent
Competing interests
Correspondence to: pain.12 Intermittent pain can be severe, and can prevent
D.A.W. D.A.W. is the holder of an Inflammation Competitive Research
david.walsh@ Programme (I-CRP) grant from Pfizer UK, and has previously acted forward planning and engagement in valued activities.
nottingham.ac.uk as a consultant for Pfizer. D.F.M. is supported by the I-CRP grant. Different mechanisms might underlie intermittent and

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  581


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points activity can also exacerbate RA fatigue,23 although the


relative importance of aerobic fitness to fatigue remains
■■ Pain in rheumatoid arthritis (RA) results from interplay between joint pathology
uncertain.24 Low mood in RA is independently associ-
and processing of pain signals by peripheral nerves, spinal and supraspinal
pain pathways ated with fatigue17 and with pain.25 Abnormalities of brain
■■ Despite good control of inflammation, pain often remains problematic in RA processing associated with fatigue and depression overlap
■■ Animal models and modern imaging techniques have illuminated the complex with those associated with pain.26
mechanisms that underlie RA pain Pain, fatigue and low mood might represent different
■■ Fatigue, stiffness and psychological distress are integral components of the RA facets of a coherent disorder of central nervous system
pain experience processing. Indeed, patients with RA are more likely to
■■ Multimodal management of RA pain includes pharmacological, psychological
satisfy classification criteria for fibromyalgia than the
and exercise-based interventions
general population.27,28 Approximately 10% of par­ticipants
with RA in one cohort satisfied ACR 2010 fibromyalgia
constant pain, with activation of mechanoreceptors criteria at any one time, and nearly 20% satis­fied fibro-
and flares of inflammatory disease being implicated in myalgia criteria during follow-up.27 RA may, therefore,
the former. The contribution of changes in central pain predispose people to developing abnormal central pain
processing to pain flares is unknown, although patients processing, as recognized in people with fibro­myalgia.
often describe worsening pain after psychological stres­ Treatments for pain, fatigue and low mood may overlap,
sors.13 Morning stiffness, a clinical feature of active syno- or even synergize.
vitis, can be perceived as part of the pain experience,14
the coincidence of the two symptoms suggesting shared Animal models of RA pain mechanisms
in­flammatory mechanisms. Pain has been widely investigated in animal models. Two
key features of RA are that pain originates from intra-
Pain, fatigue and low mood articular (rather than cutaneous) structures, and that
Fatigue is often as disabling as pain in RA; it can be medi- inflammation is chronic and immune-mediated. Mec­ha­
ated by comorbidities or psychosocial factors that are nisms of pain may differ between articular and cutaneous
common in the population, or by specific RA pathology.15 inflammation, and between acute and chronic synovitis.29
RA-associated fatigue has variously been attributed to Models of chronic immune-mediated articu­lar inflamma-
inflammatory disease activity 16 and to pain,17 although tion used to investigate pain have been induced in rats or
the association between fatigue and chronic RA pain mice using complete Freund’s adjuvant, collagen, antigen
pre­dominates.17,18 Fatigue in RA has been associated with or serum-transfer 29 (Table 2).
reported pain intensity in cross-sectional studies, and has Pain behaviour in these animal models results from
been shown to improve as pain improves.17 The associa- complex interactions between joint inflammation and
tions between pain and fatigue are independent of dis­ease altered pain processing (Table 2). Factors within the joint
activity, whereas those between fatigue and disease activ- excite or sensitize peripheral nerves, and lead to plastic-
ity could be explained by pain.17 Pain and fatigue are simi- ity of gene expression and to nerve growth. Com­plex
larly associated in fibromyalgia19 and in low back pain.20 interactions between neurons and inflammatory cells
Pain might cause fatigue by contributing to impaired sleep in the dorsal root ganglia and central nervous system
quality,21 which is commonly reported in RA. Sleep distur- lead to net facilitation or inhibition of pain signal­ling.
bance may, in turn, alter central pain processing by blunt- In some models, neuronal death may contribute to
ing descending analgesic mechanisms.22 Reduced physical neuropathic pain.29

Table 1 | Dimensions of pain examined in pain questionnaires commonly used in RA


Questionnaire RA in Severity Impact
validation
Sensory Emotional Quality Periodicity Functional Psychological Quality
population?
of life
SF-36 bodily pain Yes Yes No No No Yes No Yes
section
EQ-5D pain Yes Yes No No No No No Yes
dimension
AIMS Yes Yes Yes No No Yes Yes Yes
VAS Yes Yes No No No No No No
McGill Pain Yes Yes Yes Yes No No No No
Questionnaire
RAPS Yes Yes Yes Yes Yes Yes Yes No
WHYMPI Yes Yes Yes No No Yes Yes Yes
painDETECT No Yes No Yes Yes No No No
Abbreviations: AIMS, Arthritis Impact Measurement Scales; EQ-5D, European Quality of Life—5 dimensions; RAPS, Rheumatoid Arthritis Pain Scale; SF-36,
short-form 36; VAS, visual analogue scale; WHYMPI, West Haven–Yale Multidimensional Pain Inventory.

582  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 1 | Definitions of relevant terms* nociceptors (see Box 1). Numerous algogens (pain-­


producing agents) have been identified in the RA syno­
■■ Allodynia: pain due to a stimulus that does not normally provoke pain
vium or synovial fluids, and these factors can excite or
■■ Analgesia: absence of pain in response to stimulation which would normally
be painful sen­sitize peripheral nociceptors, often either evoking
■■ Central sensitization: increased responsiveness of nociceptive neurons in the excitatory inward currents, or modifying the function of
central nervous system to their normal or subthreshold afferent input ion channels (for example, TrpV1).33 Further­more, cyto­
■■ Hyperalgesia: increased pain from a stimulus that normally provokes pain kines (such as IL‑1β, IL‑6, and TNF), growth factors (such
■■ Neuropathic pain: pain caused by a lesion or disease of the somatosensory as β‑nerve growth factor and vascular endothelial growth
nervous system factor), and the chemokine CCL2 (also known as mono-
■■ Nociception: the neural process of encoding noxious stimuli
cyte chemotactic protein‑1) are also present at increased
■■ Nociceptive pain: pain that arises from actual or threatened damage to
non-neural tissue and is due to the activation of nociceptors
levels in RA synovial fluids,34–37 and can sensitize periph-
■■ Nociceptive stimulus: an actually or potentially tissue-damaging event eral nerves through specific cell-­surface receptors. The
transduced and encoded by nociceptors direct contribution of these factors to RA pain has been
■■ Nociceptor: a high-threshold sensory receptor of the peripheral somatosensory difficult to prove, given their pleiotropic effects and the
nervous system that is capable of transducing and encoding noxious stimuli inevitable limitations of human experimentation. Many
■■ Noxious stimulus: a stimulus that is damaging or threatens damage to proinflammatory factors can act on non-neuronal cell
normal tissues types within the joint through which they could indirectly
■■ Pain: an unpleasant sensory and emotional experience associated with actual
increase pain by stimulating the release of algesic factors.
or potential tissue damage, or described in terms of such damage
■■ Pain threshold: the minimum intensity of a stimulus that is perceived as painful Recent interest has also focused on factors—such as
■■ Peripheral sensitization: increased responsiveness and reduced threshold of endogenous opioids, somatostatin, the lipid mediators
nociceptive neurons in the periphery to the stimulation of their receptive fields known as resolvins, and anti-inflammatory cytokines
■■ Sensitization: increased responsiveness of nociceptive neurons to their normal IL‑4 and IL‑10—that may reduce neuronal sensitiv-
input, and/or recruitment of a response to normally subthreshold inputs ity.38–41 These factors have been detected in synovium and
■■ Temporal summation: an electrophysiological phenomenon of increased action synovial fluids from patients with RA,42 but their ability
potential discharge elicited by repetitive application of a noxious stimulus
to directly moderate pain from arthritic joints remains to
*With the exception of temporal summation, definitions of all terms are from the International
Association for the Study of Pain®(IASP) Taxonomy.174 This Taxonomy has been reproduced
be determined.
with permission of the IASP. The Taxonomy may not be reproduced for any other purpose
without permission. Central processing of RA pain
Several lines of evidence indicate that central pain pro-
cessing is augmented in RA (reviewed by Meeus et al.43),
Existing evidence is insufficient to determine whether as well as in OA.44 People with RA display widespread
pain mechanisms differ between monoarticular and poly­ reductions in pressure and thermal pain-thresholds and
articular arthritis models, or between those affecting increased pain sensitivity, not only over inflamed joints,
different joints. No single animal model reflects human but also at remote, nonarticular sites.45,46
disease in all respects, and data from one model might Sensitization affects a broad range of sensory modali-
not always be generalizable to other models or to human ties, in response to mechanical or thermal stimuli (see
RA.30 The translational validity of animal models of RA Box 1). In people with RA, the normal reduction of pres-
pain mechanisms is suggested by their responsiveness to sure pain thresholds that follows repeated stimulation
analgesics.31 However, lack of success in clinical trials of is augmented.47 This threshold-lowering occurs over
neurokinin‑1 receptor or purinergic receptor antagonists, a longer time course than electrophysiological tempo-
or of transient receptor potential cation channel sub­ ral summation,48 but may similarly reflect changes in
family V member 1 (TrpV1) inhibitors,32 despite promis- neuron­al adaptive responses.
ing preclinical results, suggests that mechanisms of RA Central pain processing occurs at both spinal and
pain in humans may be distinct from those observed in supra­spinal levels both in OA44 and in RA. Spread of
these rodent models. mec­hanical allodynia beyond the receptive field of cuta-
neous neurons stimulated by intradermal injection of
Mechanisms of RA pain in humans capsaicin is thought to reflect spinal pain facilitation.49,50
Despite the limitations of animal models, a picture is Res­ponses to intradermal capsaicin injection at sites dis­
emerging of peripheral and central RA pain mechanisms tant from inflamed joints in people with RA suggest that
that, in many respects, parallel those observed in animal both ipsilateral and contralateral spinal pain facilitation
models. The evidence for this view is accumulating from may occur.50
biochemical and histological studies, quantitative sensory Increased electroencephalographic activity has been
testing and neuroimaging. reported in people with RA in response to repeated pain­
ful stimuli with interstimulus intervals less than 8 s long.51
Peripheral mechanisms of RA pain Functional MRI (fMRI) enables precise anatomical delin­
Arthritis pain occurs either upon mechanical stimulation ea­tion of altered cerebral responses to painful stimuli.26
of the joint (for example, weight-bearing, appli­cation of Acutely painful stimuli applied to people with RA—for
pressure, joint movement) or spontaneously while the example, by squeezing across the meta­carpal joints of
joint is at rest. Changes in the articular environ­ment an affected hand—activate regions in the thalamus and
during RA can either activate or sensitize peripheral secondary sensory cortex, believed to reflect the sensory

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  583


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 2 | Selected details of pain mechanisms in animal models of inflammatory arthritis


Model Typical target Pain mechanisms
joints
Peripheral Spinal and supraspinal
CFA-induced Knee, TMJ, ankle, Increased innervation of the synovium140 Evidence of nociceptive pathway activity: ERK phosphorylation,144
arthritis polyarthritis Increased spontaneous and mechanically- NK‑1R internalization,145 and opioid-dependent increases in c‑Fos146
induced firing of articular primary Altered central pain processing partly mediated by increased sensitivity
afferents141 of spinal neurons via glial activation147 via IL‑1, TNF and IL‑6148,149
Increased DRG expression of Spinal neuroplasticity142 and increased expression of TrpV1,147 NK‑1R150
substance P, CGRP and NPY142 and Trk‑A in spinoreticular neurons151
Increased expression of c‑Fos143, CX3CR1 expression associated with central sensitization152
and TrpV1142, and of purinergic P2X3 Increased brain activation in the thalamus;153 increased metabolic activity
receptors32 and Trk‑A receptors for NGF140 in the anterior pretectal nucleus, anterior cingulate cortex and nucleus
accumbens;154 increased hippocampal expression of c‑Fos;155 contributions
of brainstem neurotensin and 5-hydroxytryptamine receptor156,157
Endogenous analgesia from increased vagal activity,141 opioid expression
in ganglia143 and descending inhibitory control;158 glial inactivation
converts IL‑1β facilitation into inhibition;159 DNIC augmented during acute
phase but impaired in chronic arthritis158
Collagen-induced Polyarthritis Neuronal death: few ATF3-positive DRG Spinal gliosis;29 IL‑1, TNF, and IL‑6 produced by microglia and astrocytes
arthritis cells observed29 contribute to central sensitization148,149
Glial-derived fractalkine and cathepsin S may mediate pain behaviour160
Serum transfer Polyarthritis Associated, in its later stages, with No evidence available
persistent mechanical allodynia and ATF3
expression in DRG, suggestive of sensory
nerve damage and neuropathic pain161
Antigen-induced Knee, other Mechanical hyperalgesia in ipsilateral and Spinal involvement of opioid and NMDA receptors164, TNF 165 and
arthritis single joints contralateral knees and hindpaws that is prostaglandin E2 receptor EP3 subtype166
attenuated by TNF inhibition29
Primary sensory afferents display
increased expression of pro-algesic
bradykinin receptors162
Bilaterally increased expression of NK‑1R
and B2 bradykinin receptor in DRG163
Abbreviations: ATF3, activating transcription factor 3; CFA, complete Freund’s adjuvant; CGRP, calcitonin gene-related peptide; CX3CR1, CX3C chemokine receptor 1; DNIC, diffuse noxious
inhibitory control; DRG, dorsal root ganglia; ERK, extracellular signal related kinase; NGF, nerve growth factor; NK‑1R, neurokinin‑1 receptor; NMDA, N-methyl-D-aspartate; NPY, neuropeptide Y;
TMJ, temporomandibular joint; Trk‑A, tyrosine kinase receptor A; TrpV1, transient receptor potential cation channel subfamily V member 1.

processing of pain, and also regions in the limbic system interpreted as an adaptive response to high nociceptive
(cingulate and insular cortex) that might reflect emotional input from the joints. Exposure to an acute noxious stimu-
processing.52 Furthermore, cerebral activity associated lus at a distant site reduces pain from a test site in healthy
with evoked pain in RA is moderated by psycho­logical individuals, a phenomenon described as conditioned
state.53 Activity in the medial pre­frontal cortex in response pain modulation (CPM; previously described as diffuse
to joint palpation was augmented in people who reported noxious inhibitory control). How­ever, CPM responses
more symptoms of depression, and higher depression can be normal57 or reduced in people with RA of greater
scores were associated with high tender:swollen joint than 5 years duration,59 suggesting impaired rather than
count ratios. Low mood has been associated with aug- a­ugmented endogenous central analgesic mechanisms.
mented pain processing through increased activity in Low pain thresholds in response to pressure, greater
the limbic system and descending facilitation of noci­cep­ sensitivity and reduced CPM are associated with more-
tive transmission (reviewed elsewhere54). Further­more, severe reported RA pain, indicating the clinical relevance
changes in cortical opioid-­receptor binding may underlie of quantitative sensory testing.60 However, the relative
associations between pain and mood in RA.55 contributions of nociception, peripheral sensitization,
As well as these changes in functional activity, the central facilitation and loss of central inhibition to RA
brains of people with chronic pain can display altered pain remain uncertain, and are likely to vary from person
structure. Basal ganglia grey matter volume was increased to person, and over time. Furthermore, available data are
in people with RA compared with pain-free individuals, insufficient to characterize differences between RA and
although longitudinal studies would be needed to deter- other forms of chronic musculoskeletal pain.
mine whether this difference in brain volume represents
an adaptive change to chronic-pain processing.56 Psychological aspects of RA pain
Under some circumstances, people with RA report less Pain exacerbates psychological distress, and stress can
pain in response to a standardized stimulus than do healthy augment pain. Patients with RA report worse mental
individuals, suggesting hypoaesthesia.57 Brain activity in health, with higher levels of depression and anxiety, than
response to an acute, painful, thermal stimulus may be do healthy individuals.61–63 Depression can mediate the
reduced in people with RA as demonstrated by PET,58 association between pain and impaired cognitive function

584  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

in RA,64 which further contributes to disability and dis- they initiate and facilitate both acute and chronic ‘neuro­
tress. Pain is associated with current psychological di­stress genic’ inflammation85 (this topic is comprehensive­ly
in both early and late RA.62 Depression and anxiety are reviewed elsewhere86).
inter-related constructs, and both may contribute to RA Acute painful stimuli activate neuroendocrine res­
pain.65 Pain predicts incident depression in RA,66 and ponses, which may be either anti-inflammatory or pro-
people with a history of depression have reported worse inflammatory, both in people with and without RA.45,87
RA pain than those with no such history,67 particularly Pain stimulates increased production not only of gluco-
if they have suffered repeated depressive episodes.68 corticoids, but also of proinflammatory cytokines includ-
Although previous depression was not predictive of worse ing IL‑6 and TNF.33 Circulating levels of TNF increase
pain in all studies,69 psychological characteristics can after induction of experimental pain in people with RA,
predict pain better than do measures of joint structure or whereas anti-inflammatory cortisol responses are nor­
inflammation.70 Patients report pain as the major barrier mal45 or even blunted.88 Changes in neuroendocrine
to their psychological well-being, and the effects of active res­ponses to painful stimuli have been observed in early
inflammation upon mental distress seem to be mediated RA,89 but it is uncertain whether an unfavourable balance
by pain.71 Improvements in pain are associated with reduc- of neuroendocrine responses represents a predisposing
tions in distress.72 Furthermore, people with a history of phenotype or is a mediator of augmented-pain pr­ocessing
depression may be more susceptible to mood disturbance or synovitis.
associated with RA pain than those without.69 Other forms of stress can indirectly affect pain by aug-
Both in healthy people and in those with RA, acute menting inflammatory disease activity in RA,45 through
and chronic psychological stressors can augment central neuroendocrine pathways90 such as the h­ypothalamic–­
pain processing.62,73 Pain thresholds in RA are particu- pituitary–adrenal axis and the sympathetic–­adreno­
larly reduced in people who report low mood, even in the medullary system.91 Chronic stress can increase circu­lating
absence of clinical depression.74 Psychological or pharma- levels of proinflammatory cytokines and C-reactive pro­
cological treatments that reduce distress can alter central tein,92 and severe psychological stress has been p­roposed
pain processing,75 and reduce pain,76 in people with RA. as a trigger for RA onset.93
Unhelpful beliefs about the nature, meaning and
adverse consequences of pain can contribute to reported Pain progression in RA
pain intensity and psychological distress and be a barrier Traditional models of RA, in which symptoms in early
to effective treatment.77 For example, a higher number of disease are attributable to inflammation whereas those
reasons given by people with RA why they should not to in late disease are attributable to inflammation and also
take medication was associated with worse pain.78 As in joint damage, require substantial modification in order
other chronic pain conditions, catastrophizing has been to reflect the complex mechanisms that underlie RA
associated with worse RA pain.79 pain (Figure 1). Genetic constitution,94 comorbid condi-
Unhelpful beliefs and low mood can lead people with tions95 and augmented central pain processing before RA
RA to adopt ineffective coping strategies and to experi- onset may influence subsequent RA pain. Inflamma­tion
ence a sense of helplessness and reduced self-efficacy. is followed by rapid peripheral and central sensitiza­tion.
Passive coping, helplessness and low self-efficacy or per- Psycho­logical aspects of RA pain similarly evolve, with
sonal control over pain are associated with worse pain changes in mood96 and acceptance over the course of
and depression in RA.69,80 Passive coping may mediate the the disease. Structural causes of RA pain are fea­tures
association between pain and physical disability in RA.81 of later disease, although subchondral inflammation
Conversely, high levels of self-efficacy for pain could help might contribute to pain in even early RA. Altered central
people with RA to achieve their physical function goals, pain processing across multiple pain modalities induced
in order to improve their quality of life.82 by mechanical or thermal stimuli may be particularly
Several studies have addressed the adverse influence characterist­ic of patients with longstanding disease.57
of social stressors, such as difficult marital partnerships, Predictors of poor pain outcomes in RA include poor
on RA pain.83 Patient reports of negative spousal behav- mental health97 and evidence of abnormal central pain pro-
iour (such as avoidance and critical remarks) predicted cessing.95,98 Mechanisms that contribute to pain progres-
worse RA pain.84 Conversely, daily emotional support sion therefore differ from those that contribute to persistent
and social life domains associated with positive affect inflammatory disease or progressive structural damage.
improved pain outcomes.84 Specifically, spousal support
seemed to reduce the negative effects of catastrophizing Management of RA pain
on RA pain.79 Pain in RA is a multidimensional experience, resulting
from multiple mechanisms, and its management there-
Effects of pain on inflammation fore requires not only detailed assessment of its causes
The potential for both the peripheral and central nervous and its effects on the individual patient, but also coordi­
systems to influence inflammation is well documented, nated provision of pharmacological, psychological and
although the importance of this influence in RA remains physio­therapeutic approaches. A range of evidence-based
incompletely understood. Peripheral sensory nerve activa- treatments, discussed in this section, can complement
tion leads to release of neuropeptides, such as sub­stance P each other and could be used in combination to try and
and calcitonin gene-related peptide, into the joint, where m­inimize the impact of pain on individuals with RA.

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  585


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Pathological mechanisms contributing to pain effects reported even with paracetamol, particularly
Chronic inflammation when used in conjunction with NSAIDs.101 As with all
Risk factors treatments, analgesics should only be continued when
for pathology Psychological distress
Genetics there is benefit for the individual that outweighs current
(e.g. HLA-DR) Structural change or potential adverse events. Placebo effects and regres-
Comorbidities
(e.g. OA, sion to the mean reportedly contribute approximately
Acute inflammatory flares
depression) half of analgesic efficacy in RCTs in OA102 and in RCTs
of analge­sic treatments in RA,103,104 and should be con-
Pre-morbid Early RA Established RA
sidered when reviewing individual responses to analgesia.
The relative values placed on risks and benefits often vary
Risk factors for pain
between individuals, and analgesic use should be based
Genetics Altered central pain processing
Comorbidities Sensitization (spinal and supraspinal), on informed choices made by people with RA. However,
(e.g. fibromyalgia) descending facilitation and inhibition despite these limitations, directly acting analgesics could
Psychological
context provide substantial and rapid benefit to people with RA.
(e.g. mood, beliefs) Nociceptor activation and peripheral sensitization
Pain mechanisms contributing to RA experience Reducing pain by suppressing inflammation
Treatments that target synovial inflammation reduce
Figure 1 | Pain mechanisms through the natural history of RA. Diagram representing
the time course from pre-morbid (left) through early and established RA (right).
RA pain. Glucocorticoids, delivered by intra-­articular,105
Different pain mechanisms throughout the natural history of RA are shown. Green oral106 or parenteral107 routes, may provide clinically
shading represents pathological changes, related to both the joint and potential meaningful pain relief in RA through anti-­inflammatory
psychopathology (for example, depression). Purple shading represents pain mechanisms. Pain reduction can be due to local or sys-
processing. Red shading represents the self-limiting acute flares that contribute to temic actions108 and, although symptoms may rebound
RA pain. Comorbidities that precede RA onset may predispose individuals to worse after steroids are discontinued, continued benefit has been
pain. Genetic variation may predispose individuals to synovitis, psychopathology and demonstrated for up to 1 year with system­ic treatment.109
joint damage, and also may influence pain processing following RA onset. Changes
Traditional DMARDs such as methotrexate, sulphasala-
in pain processing occur sequentially after the onset of inflammation, with rapid
peripheral sensitization followed by microglial then astrocyte activation in the spinal zine and leflunomide also reduce joint pain,110–112 with an
cord, altered gene expression, sustained changes in brain activity and structure. onset of action that parallels suppression of inflamma­tion
Psychological factors, such as low mood or unhelpful beliefs, can also evolve over several weeks, and can be maintained over months.
during the course of RA. Structural causes of pain are features of later disease. Current guidelines recommend rapid introduction of
Abbreviations: OA, osteoarthritis; RA, rheumatoid arthritis. combination DMARD therapy, including metho­trexate
and glucocorticoids. 113 Combination therapy can be
Pharmacological management of RA pain su­perior to monotherapies for pain reduction, with com-
Pharmacological treatments that improve RA pain bination therapy associated with long-term pain scores
include directly acting analgesic agents, drugs that sup- that were comparable to the Dutch national average in the
press inflammation, and strategies to reduce joint damage. BeSt study.114 Pain from active disease, when inade­quately
Pain outcomes in RA can be poor despite optimal control controlled by metho­trexate, can also be improved further
of inflammatory disease; 4 in one study of early RA, by addition of a biologic therapy,113 with a 60% reduction
12 months after diagnosis and DMARD initiation pain in reported pain (compared with 29% after the addition
remained a major problem.95 Therefore, directly acting of placebo) in one trial.115 Benefits in routine clinical prac-
analgesics are recommended, concurrent with strategies tice, however, may not match those observed in clinical
aimed at controlling inflammatory disease. trials, possibly reflecting less highly selected patient popu-
lations, and greater difficulty in achieving and maintain-
Directly acting analgesic agents ing optimal treatment. In a UK inception cohort study
A large number of analgesic drugs have the potential to of early RA (the Early RA Network), patients commenc-
improve pain, although their use is rarely supported by ing treatment with nonbiologic DMARDs showed a 20%
randomized controlled trials (RCTs) in RA (Table 3). improvement in short-form 36 bodily pain scores within
Furthermore, analgesic trials in RA are typically of short the first 12 months,95 which then maintained a plateau
duration, and evidence of long-term efficacy beyond for the next 4 years that remained worse than the UK
6 weeks is weak.99 Large trials that minimize risk of bias are mean. Pain may, therefore, remain problematic despite
required to define the optimal use of analgesics in people treatment with conventional DMARDs.
with RA. Biologic agents, including TNF blockers, anakinra, toci-
Characterization of RA pain mechanisms, comorbidi- lizumab and rituximab, reduce joint pain in RA, usually
ties and concurrent treatments informs the selection of within 2–4 weeks of their initiation, in parallel with reduc-
treatments that have the most favourable risk:benefit ratios tions in synovitis. The main mechanisms for improving
for the individual. The balance between peripheral and pain are thought to be reduction of inflammation,116
central pain mechanisms may be especially important. dampening peripheral and central sensitization117,118 and
For example, NSAIDs improve pain in RA, but have little long-term prevention of joint damage.116,119 Some patients
benefit as monotherapy for people with centrally mediated report pain relief within 24 h of TNF-blocker administra-
pain, such as fibromyalgia.100 Adverse events frequently tion, well before demonstrable effects on inflammation.
limit analgesic use, with substantial gastrointestinal toxic Brain activation and functional connectivity decreased

586  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 3 | Evidence for pharmacological analgesic efficacy in RA* structural changes that directly mediate pain, and can have
better predictive value for measuring joint pain.125
Analgesic Summary of evidence in RA Reference(s)
agent clinical trials Effective suppression of inflammatory disease reduces
progression of erosive damage126 which might mediate
Paracetamol 12 RCTs and one observational study in RA Hazlewood et al.
showed weak evidence of benefit over (2012)167 permanent reductions in pain. Intensive treatment of early
placebo and an additive benefit when used RA with glucocorticoids and DMARDs or biologic agents
together with NSAIDs reduces structural progression and results in long-term
NSAIDs Weak evidence shows that physicians and Wienecke et al. (2004)168 pain-reduction.114 Delayed introduction of DMARDs is
patients prefer ibuprofen over paracetamol McCormack (2011)169 associated with worse pain 12 months after presentation.99
COX-2-selective and nonselective inhibitors Chen et al. (2008)170
Improved pain outcomes with early treatment might result
seem to be equally efficacious in RA
from sustained suppression of inflammation, or from
Opioids 11 studies in RA, where four could be Whittle et al. (2012)171
reducing the development of central pain augmentation,
pooled to show opioids led to improved
pain relief over placebo, but with an as well as from reducing structural damage.127
increased risk of AEs; high likelihood of bias
Tricyclic Nine RCTs in inflammatory arthritis Richards et al. (2012)172 Psychological management of RA pain
antidepressants (eight in RA) found no evidence of Unfortunately, complete abrogation of pain is not a real-
short-term (<1 week) benefit istic goal for many people with RA at present. Patients
Conflicting evidence found in trials of
duration >1 week, with minor AEs that
in clinical remission of inflammatory disease commonly
did not result in discontinuation have persistent pain,4 and relapse of inflammatory disease
Nefopam Two trials with risk of bias showed benefit Richards et al. (2012)173
is not uncommon.128 People with RA live with their pain,
of nefopam over placebo, but with and develop psychological strategies that may either help
associated AEs or hinder their ability to lead fulfilling lives. Psychological
Capsaicin One trial found topical capsaicin was Richards et al. (2012)173 interventions such as cognitive behavioural therapy (CBT)
beneficial over placebo, but with some AEs aim to improve well-being in the setting of chronic or inter-
*Agents listed may not be licensed for use for RA pain in some countries. Abbreviations: AE, adverse mittent pain. CBT improves disability and has small effects
event; COX, cylooxygenase; RA, rheumatoid arthritis; RCT, randomized controlled trial.
on reported pain severity.76,129 A 2010 meta-analysis of 27
trials of face-to-face psychological interventions, incor-
in parallel to this rapid analgesia,120 and brain responses porating CBT (10 trials), education (nine trials) or stress
to pressure across the metacarpophalangeal joints were management (two trials), in RA indicated small effects
reduced when assessed by fMRI, particularly in regions on pain reduction (pooled effect sizes 0.18 immediately
associated with the sensory (thalamus, somatosensory post-treatment, 0.13 at 2–14 month follow-up).130
cortex) and emotional (cingulate and insular cortex) pro- CBT focuses on changing unhelpful beliefs, encourag-
cessing of pain.52,120 These data raise the possibility that ing productive coping strategies and facilitating behav-
clinical response to TNF-blockers may partially depend ioural change. Alternative psychological approaches,
on moderation of cerebral function, perhaps involving such as mindfulness training and acceptance and com-
descending control of nociceptive inputs and emotional mitment therapy (ACT), may also benefit people with
processing of pain. However, these studies were not able RA.76 Mindfulness and ACT aim to prevent adverse
to distinguish between direct supraspinal actions and their con­se­­quences without necessarily changing beliefs. An
effects mediated by reductions in peripheral noci­ceptive RCT com­paring CBT with mindfulness training or an
input. Placebo effects might also contribute to rapid ed­ucation-based control suggested that all interventions
analge­sia after TNF-blocker administration, although were associated with similarly small reductions in reported
simi­larly rapid reduction of pain behaviour has been pain intensity;76 however, CBT and mindfulness training
observed in mice with collagen-induced arthritis.52 had greater benefits on the psychological consequences of
pain. CBT was more beneficial for reported pain-control
Structural damage and management of RA pain than was mindfulness training, whereas mindfulness
The contribution of structural changes within the joint training seemed to be helpful for pain-coping and catas­
to RA pain remains controversial. Radiographic erosions trophizing, particularly in people with a history of depres-
and joint-space narrowing are associated with disability sion. Mindfulness and CBT could, therefore, be suited to
in patients with longstanding RA,121 and make a small but different client groups, based on history of depression.
statistically significant contribution to reported RA pain, Previous or current depression may not only augment RA
as do psychological factors and synovitis.122 Joint-space pain, but may also predict response to different treatments.
narrowing also indicates that comorbid or secondary OA The potential power of psychological factors to influence
could contribute to pain in people with RA. However, pain is highlighted by the magnitude of placebo effects in
radiographic scores may contribute only a small propor- RCTs. Effect sizes for placebos in OA, compared with base-
tion to reported symptoms after adjustment for traditional line or usual care, are of the order 0.4–0.5 (moderate effect
measures of inflammatory disease activity.123 Benefits of size),102 and comparable to the incremental effect sizes
total joint replacement for people with RA might exceed of most pharmacological analgesics. Placebo effects are
those even in OA,124 suggesting the importance of struc- dependent on the context in which treatments are given,
tural mediators of pain in established RA. Imaging tools both the internal context (for example, psychological dis-
other than radiography, such as MRI, might better detect tress, beliefs, genes), and the external context (for example,

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  587


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

interactions between patient and health-care professional, could avoid some problems with composite measures,
mode of treatment delivery). Enhancing the context of but might not adequately reflect the patient’s experience.
treatment delivery in RA might be almost as important as Acute-phase responses could reflect comorbid disease,
developing novel pharmacological treatments in improving swollen joint counts might be influenced by interactions
patient well-being. between the patient and clinician, and ultrasound evi-
dence of synovitis can be present even in asymptomatic
Exercise as a treatment for RA pain joints.139 Objective measures of synovitis require validation
Although exercising an arthritic joint can be painful, against patient-centred outcomes, in order that they can
several lines of evidence indicate that exercise modifies be incorporated into evidence-based guidelines for RA
central pain processing, and therefore reduces pain. In management. Meanwhile, clinicians should be aware of the
nonarthritic populations, aerobic exercise can increase potential pitfalls of assuming all pain in RA is attributable
pain-detection and tolerance thresholds, and reduces to inflammation, or indeed that improvements in pain are
reported pain intensity in response to a nociceptive stimu- attributable to reduced inflammation.
lus.131 The mechanisms behind acute exercise-induced
analgesia are incompletely understood, but might include Conclusions and future directions
increased levels of endorphins.132 In addition, aerobic Pain remains the most important symptom for many
fitness training may be associated with sustained increases people with RA, despite recent advances in disease-­
in circulating endorphin levels132 and inhibition of spinal modifying treatments. Inflammation, peripheral and
nociceptive reflexes.133 central pain processing, and structural change within
Exercise-induced analgesia might depend on the the joint each contribute to RA pain. RA pain may be
intensity and type of exercise. RCTs of aerobic exercise constant or intermittent, localized or widespread, and is
in people with RA, in whom the inflammatory com- commonly associated with psychological distress and
ponent of their disease is under stable control, indicate fatigue. Dominant pain mechanisms in an individual
weak but clinically important reductions in reported pain are identified by critical evaluation of clinical symptoms
(effect size approximately 0.3134), although the effects of and signs, laboratory tests and imaging, although further
resistance exercises on RA pain did not reach statistical research is required to define quantitative tools in order to
significance.134 Furthermore, exercise may have differ- inform consistent, evidence-based treatment stratification.
ent effects according to the underlying diagnosis. T’ai Fibromyalgia and OA are not diagnoses of exclusion in RA,
Chi has encour­aging effects on pain in people with OA, but instead, pain mechanisms common to these conditions
whereas clinical trials in RA have not yet confirmed an and RA may require treatment in paral­lel to the manage-
important analge­sic benefit.135 People with fibromyalgia ment of inflammatory disease. Pain is an important com-
may experi­ence reduced, rather than increased, pain thres­ ponent in the clinical assessment of inflammatory disease
holds during exercise,136 perhaps indicating impairment of activity, and non­inflammatory components of RA pain
descending inhibitory control. should be considered when gauging the appropriate­ness
of or anti-­inflammatory response to treatments. Synovitis
Pain and assessment of disease activity may initiate or maintain abnormalities in central pain pro-
Inflammatory disease activity is assessed in clinical prac- cessing, and already some evidence shows that effective
tice and in research using composite scoring methods suppression of inflammatory disease might help no­rmalize
such as the 28-joint disease activity score (DAS28). These pain processing.
scores combine patient-reported with observed and A range of pharmacological analgesics, immuno­modu­
la­boratory-measured components.137 Patient-reported latory agents, psychological interventions, exercise and
outcomes in RA are strongly influenced by pain. Pain can sur­gery are available to help people with RA pain. More
be an important indicator of active inflammation, but RCTs are required to determine the efficacy and optimal
non­inflammatory mechanisms can also importantly con- usage of analgesics in RA, and novel analgesic agents with
tribute to pain, and may confound assessments of disease greater efficacy and lower propensity for adverse reactions
activity. Furthermore, concurrent analgesia could poten- are urgently needed. In the meantime, more judicious use
tially mask or augment evidence of inflammatory disease of existing treatments has the potential to improve the lives
response based on DAS28. of individuals with RA pain.
DAS28‑P, which describes the proportion of DAS28
score attributable to patient-reported components (tender Review criteria
joint count and visual analogue scale score of general Articles for this Review were chosen from the authors’
health), has been proposed as a surrogate measure of personal libraries and from searches of Ovid MEDLINE
non­inflammatory pain mechanisms in people with RA.95 using the search terms “rheumatoid arthritis”, “pain”,
DAS28‑P is associated with pain, and predicts pain out- “randomised controlled trial”, “experimental arthritis”,
“central sensitisation”, “glial”, “astrocyte”, “spine”,
comes in prospective cohorts. High ratios of tender:swollen “inflammation”, “synovitis”, “psychology”, “depression”,
joint counts may similarly indicate non­inflammatory “catastrophisation”, “CBT”, “anxiety”, “physiotherapy”,
pain mechanisms.98 “exercise”, “validation”, “questionnaire” and other
Inflammatory disease assessment using objective meas- related terms. Full-length papers were retrieved and the
ures, such as biomarkers of acute-phase response, swollen authors’ expert opinions determined their inclusion.
Searches were performed up to December 2013.
joint counts95 or ultrasonographic evidence of synovitis,138

588  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Merskey, H. & Bogduk, N. (eds) Classification patients with rheumatoid arthritis. Pain 153, of inflammatory arthritis and may predict
of Chronic Pain (IASP Publications, Seattle, 107–112 (2012). response to therapy. Rheumatology (Oxford) 51,
1994). 22. Lee, Y. C. et al. The role of sleep problems in 451–459 (2012).
2. Heiberg, T. & Kvien, T. K. Preferences for central pain processing in rheumatoid arthritis. 41. Xu, Z. Z. et al. Resolvins RvE1 and RvD1 attenuate
improved health examined in 1,024 patients Arthritis Rheum. 65, 59–68 (2013). inflammatory pain via central and peripheral
with rheumatoid arthritis: pain has highest 23. Scarvell, J. & Elkins, M. R. Aerobic exercise is actions. Nat. Med. 16, 592–597 (2010).
priority. Arthritis Rheum. 47, 391–397 (2002). beneficial for people with rheumatoid arthritis. 42. Giera, M. et al. Lipid and lipid mediator profiling
3. da Rocha Castelar Pinheiro, G., Khandker, R. K., Br. J. Sports Med. 45, 1008–1009 (2011). of human synovial fluid in rheumatoid arthritis
Sato, R., Rose, A. & Piercy, J. Impact of 24. Munsterman, T., Takken, T. & Wittink, H. patients by means of LC-MS/MS. Biochim.
rheumatoid arthritis on quality of life, work Low aerobic capacity and physical activity not Biophys. Acta 1821, 1415–1424 (2012).
productivity and resource utilisation: an associated with fatigue in patients with 43. Meeus, M. et al. Central sensitization in patients
observational, cross-sectional study in Brazil. rheumatoid arthritis: a cross-sectional study. with rheumatoid arthritis: a systematic literature
Clin. Exp. Rheumatol. 31, 334–340 (2013). J. Rehabil. Med. 45, 164–169 (2013). review. Semin. Arthritis. Rheum. 41, 556–567
4. Lee, Y. C. et al. Pain persists in DAS28 25. Edwards, R. R., Cahalan, C., Mensing, G., (2012).
rheumatoid arthritis remission but not in ACR/ Smith, M. & Haythornthwaite, J. A. Pain, 44. Malfait, A.-M. & Schnitzer, T. J. Towards a
EULAR remission: a longitudinal observational catastrophizing, and depression in the mechanism-based approach to pain
study. Arthritis Res. Ther. 13, R83 (2011). rheumatic diseases. Nat. Rev. Rheumatol. 7, management in osteoarthritis. Nat. Rev.
5. Burckhardt, C. S. The use of the McGill Pain 216–224 (2011). Rheumatol. 9, 654–664 (2013).
Questionnaire in assessing arthritis pain. 26. Jones, A. K., Huneke, N. T., Lloyd, D. M., 45. Edwards, R. R. et al. Enhanced reactivity to pain
Pain 19, 305–314 (1984). Brown, C. A. & Watson, A. Role of functional in patients with rheumatoid arthritis. Arthritis
6. Charter, R. A., Nehemkis, A. M., Keenan, M. A., brain imaging in understanding rheumatic pain. Res. Ther. 11, R61 (2009).
Person, D. & Prete, P. E. The nature of arthritis Curr. Rheumatol. Rep. 14, 557–567 (2012). 46. Pollard, L. C., Ibrahim, F., Choy, E. H. &
pain. Br. J. Rheumatol. 24, 53–60 (1985). 27. Wolfe, F., Hauser, W., Hassett, A. L., Katz, R. S. Scott, D. L. Pain thresholds in rheumatoid
7. Roche, P. A., Klestov, A. C. & Heim, H. M. & Walitt, B. T. The development of fibromyalgia-I: arthritis: the effect of tender point counts and
Description of stable pain in rheumatoid examination of rates and predictors in patients disease duration. J. Rheumatol. 39, 28–31
arthritis: a 6 year study. J. Rheumatol. 30, with rheumatoid arthritis (RA). Pain 152, (2012).
1733–1738 (2003). 291–299 (2011). 47. Wendler, J. et al. Patients with rheumatoid
8. Harden, R. N. Chronic neuropathic pain. 28. Lee, Y. C. et al. Incidence and predictors of arthritis adapt differently to repetitive painful
Mechanisms, diagnosis, and treatment. secondary fibromyalgia in an early arthritis stimuli compared to healthy controls. J. Clin.
Neurologist 11, 111–122 (2005). cohort. Ann. Rheum. Dis. 72, 949–954 (2013). Neurosci. 8, 272–277 (2001).
9. Perrot, S., Dieude, P., Perocheau, D. & 29. Schaible, H.-G. & Brauer, R. in Pain Models: 48. Arendt-Nielsen, L. & Petersen-Felix, S. Wind-up
Allanore, Y. Comparison of pain, pain burden, Translational Relevance and Applications (eds and neuroplasticity: is there a correlation to
coping strategies, and attitudes between Handwerker, H. O. & Arendt-Nielsen, L.) 149–162 clinical pain? Eur. J. Anaesthesiol. Suppl. 10, 1–7
patients with systemic sclerosis and patients (IASP Press, 2013). (1995).
with rheumatoid arthritis: a cross-sectional 30. Vincent, T. L., Williams, R. O., Maciewicz, R., 49. Morris, V. H., Cruwys, S. C. & Kidd, B. L.
study. Pain Med. 14, 1776–1785 (2013). Silman, A. & Garside, P. Mapping pathogenesis Characterisation of capsaicin-induced
10. Cook, D. B. et al. Functional imaging of pain in of arthritis through small animal models. mechanical hyperalgesia as a marker for altered
patients with primary fibromyalgia. J. Rheumatol. Rheumatology (Oxford) 51, 1931–1941 (2012). nociceptive processing in patients with
31, 364–378 (2004). 31. McQueen, D. S., Iggo, A., Birrell, G. J. & rheumatoid arthritis. Pain 71, 179–186 (1997).
11. Hochman, J. R., Davis, A. M., Elkayam, J., Grubb, B. D. Effects of paracetamol and aspirin 50. Shenker, N. G., Haigh, R. C., Mapp, P. I.,
Gagliese, L. & Hawker, G. A. Neuropathic pain on neural activity of joint mechanonociceptors in Harris, N. & Blake, D. R. Contralateral
symptoms on the modified painDETECT adjuvant arthritis. Br. J. Pharmacol. 104, hyperalgesia and allodynia following intradermal
correlate with signs of central sensitization in 178–182 (1991). capsaicin injection in man. Rheumatology
knee osteoarthritis. Osteoarthritis Cartilage 21, 32. Shinoda, M., Ozaki, N., Asai, H., Nagamine, K. & (Oxford) 47, 1417–1421 (2008).
1236–1242 (2013). Sugiura, Y. Changes in P2X3 receptor expression 51. Hummel, T., Schiessl, C., Wendler, J. & Kobal, G.
12. Mengshoel, A. M. & Forre, O. Pain and fatigue in the trigeminal ganglion following monoarthritis Peripheral and central nervous changes in
in patients with rheumatic disorders. Clin. of the temporomandibular joint in rats. Pain 116, patients with rheumatoid arthritis in response
Rheumatol. 12, 515–521 (1993). 42–51 (2005). to repetitive painful stimulation. Int. J.
13. Zautra, A. J., Hamilton, N. A., Potter, P. & Smith, B. 33. Shubayev, V. I., Kato, K. & Myers, R. E. in Psychophysiol. 37, 177–183 (2000).
Field research on the relationship between stress Translational Pain Research: from Mouse to Man 52. Hess, A. et al. Blockade of TNF-α rapidly inhibits
and disease activity in rheumatoid arthritis. (eds Kruger, L. & Light, A. R.) (CRC Press, 2010). pain responses in the central nervous system.
Ann. NY Acad. Sci. 876, 397–412 (1999). 34. Brennan, F. M. & McInnes, I. B. Evidence that Proc. Natl Acad. Sci. USA 108, 3731–3736
14. Lems, W. F. & Dijkmans, B. A. C. in Rheumatoid cytokines play a role in rheumatoid arthritis. (2011).
Arthritis: New Frontiers in Pathogenesis and J. Clin. Invest. 118, 3537–3545 (2008). 53. Schweinhardt, P. et al. Investigation into the
Treatment (eds Firestein, G. S., Panayi, G. S. & 35. Scuri, M., Samsell, L. & Piedimonte, G. The role neural correlates of emotional augmentation of
Wollheim, F. A.) 213–225 (OUP, Oxford, 2000). of neurotrophins in inflammation and allergy. clinical pain. Neuroimage 40, 759–766 (2008).
15. Wolfe, F., Hawley, D. J. & Wilson, K. The Inflamm. Allergy Drug Targets 9, 173–180 54. Robinson, M. J. et al. Depression and pain.
prevalence and meaning of fatigue in rheumatic (2010). Front. Biosci. (Landmark Ed.) 14, 5031–5051
disease. J. Rheumatol. 23, 1407–1417 (1996). 36. Antonelli, A. et al. Chemokine (C-X-C motif) ligand (2009).
16. Crosby, L. J. Factors which contribute to fatigue (CXCL)10 in autoimmune diseases. Autoimmun. 55. Jones, A. K. et al. Endogenous opiate response
associated with rheumatoid arthritis. J. Adv. Rev. 13, 272–280 (2014). to pain in rheumatoid arthritis and cortical and
Nurs. 16, 974–981 (1991). 37. Fava, R. A. et al. Vascular permeability factor/ subcortical response to pain in normal
17. Pollard, L. C., Choy, E. H., Gonzalez, J., endothelial growth factor (VPF/VEGF): volunteers using positron emission tomography.
Khoshaba, B. & Scott, D. L. Fatigue in accumulation and expression in human synovial Int. J. Clin. Pharmacol. Res. 11, 261–266 (1991).
rheumatoid arthritis reflects pain, not disease fluids and rheumatoid synovial tissue. J. Exp. 56. Wartolowska, K. et al. Structural changes of the
activity. Rheumatology (Oxford) 45, 885–889 Med. 180, 341–346 (1994). brain in rheumatoid arthritis. Arthritis Rheum.
(2006). 38. Toth, K. et al. Synovial fluid β-endorphin level in 64, 371–379 (2012).
18. Belza, B. L. Comparison of self-reported fatigue avascular necrosis, rheumatoid arthritis, and 57. Leffler, A. S., Kosek, E., Lerndal, T., Nordmark, B.
in rheumatoid arthritis and controls. osteoarthritis of the femoral head and knee. & Hansson, P. Somatosensory perception and
J. Rheumatol. 22, 639–643 (1995). A controlled pilot study. Clin. Rheumatol. 30, function of diffuse noxious inhibitory controls
19. Wolfe, F. Fibromyalgia: the clinical syndrome. 537–540 (2011). (DNIC) in patients suffering from rheumatoid
Rheum. Dis. Clin. North Am. 15, 1–18 (1989). 39. Denko, C. W. & Malemud, C. J. Role of the arthritis. Eur. J. Pain 6, 161–176 (2002).
20. Feuerstein, M., Carter, R. L. & Papciak, A. S. growth hormone/insulin-like growth factor-1 58. Jones, A. K. & Derbyshire, S. W. Reduced
A prospective analysis of stress and fatigue in paracrine axis in rheumatic diseases. Semin. cortical responses to noxious heat in patients
recurrent low back pain. Pain 31, 333–344 Arthritis Rheum. 35, 24–34 (2005). with rheumatoid arthritis. Ann. Rheum. Dis. 56,
(1987). 40. Wright, H. L., Bucknall, R. C., Moots, R. J. & 601–607 (1997).
21. Nicassio, P. M. et al. The contribution of pain and Edwards, S. W. Analysis of SF and plasma 59. Meeus, M. et al. Does acetaminophen activate
depression to self-reported sleep disturbance in cytokines provides insights into the mechanisms endogenous pain inhibition in chronic fatigue

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  589


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

syndrome/fibromyalgia and rheumatoid 77. Edwards, R. R. et al. Moderators of the negative evidence for involvement of the 5p15.2 region.
arthritis? A double-blind randomized controlled effects of catastrophizing in arthritis. Pain Med. Ann. Rheum. Dis. 72, 427–436 (2013).
cross-over trial. Pain Physician 16, E61–E70 11, 591–599 (2010). 95. McWilliams, D. F. et al. Predictors of change
(2013). 78. Fitzcharles, M.-A., DaCosta, D., Ware, M. A. & in bodily pain in early rheumatoid arthritis:
60. Lee, Y. C. et al. The role of sleep problems in Shir, Y. Patient barriers to pain management may an inception cohort study. Arthritis Care Res.
central pain processing in rheumatoid arthritis. contribute to poor pain control in rheumatoid (Hoboken) 64, 1505–1513 (2012).
Arthritis Rheum. 65, 59–68 (2013). arthritis. J. Pain 10, 300–305 (2009). 96. Sharpe, L., Sensky, T. & Allard, S. The course of
61. Edwards, R. R., Bingham, C. O. 3rd, Bathon, J. & 79. Holtzman, S. & Delongis, A. One day at a time: depression in recent onset rheumatoid arthritis:
Haythornthwaite, J. A. Catastrophizing and pain the impact of daily satisfaction with spouse the predictive role of disability, illness perceptions,
in arthritis, fibromyalgia, and other rheumatic responses on pain, negative affect and pain and coping. J. Psychosom. Res. 51, 713–719
diseases. Arthritis Rheum. 55, 325–332 (2006). catastrophizing among individuals with (2001).
62. Edwards, R. R., Cahalan, C., Mensing, G., rheumatoid arthritis. Pain 131, 202–213 97. Odegard, S., Finset, A., Mowinckel, P., Kvien, T. K.
Smith, M. & Haythornthwaite, J. A. Pain, (2007). & Uhlig, T. Pain and psychological health status
catastrophizing, and depression in the rheumatic 80. Covic, T., Adamson, B., Spencer, D. & Howe, G. over a 10-year period in patients with recent
diseases. Nat. Rev. Rheumatol. 7, 216–224 A biopsychosocial model of pain and depression onset rheumatoid arthritis. Ann. Rheum. Dis. 66,
(2011). in rheumatoid arthritis: a 12-month longitudinal 1195–1201 (2007).
63. Goldenberg, D. L. The interface of pain and study. Rheumatology (Oxford) 42, 1287–1294 98. Pollard, L. C., Kingsley, G. H., Choy, E. H. &
mood disturbances in the rheumatic diseases. (2003). Scott, D. L. Fibromyalgic rheumatoid arthritis
Semin. Arthritis Rheum. 40, 15–31 (2010). 81. Covic, T., Adamson, B. & Hough, M. The impact and disease assessment. Rheumatology
64. Brown, S. C., Glass, J. M. & Park, D. C. The of passive coping on rheumatoid arthritis pain. (Oxford) 49, 924–928 (2010).
relationship of pain and depression to cognitive Rheumatology (Oxford) 39, 1027–1030 (2000). 99. Walsh, D. A. & McWilliams, D. F. Pain in
function in rheumatoid arthritis patients. Pain 82. Knittle, K. P. et al. Effect of self-efficacy and rheumatoid arthritis. Curr. Pain Headache Rep.
96, 279–284 (2002). physical activity goal achievement on arthritis 16, 509–517 (2012).
65. Odegard, S., Finset, A., Mowinckel, P., Kvien, T. K. pain and quality of life in patients with 100. Kroenke, K., Krebs, E. E. & Bair, M. J.
& Uhlig, T. Pain and psychological health status rheumatoid arthritis. Arthritis Care Res. Pharmacotherapy of chronic pain: a synthesis
over a 10-year period in patients with recent (Hoboken) 63, 1613–1619 (2011). of recommendations from systematic reviews.
onset rheumatoid arthritis. Ann. Rheum. Dis. 66, 83. Reese, J. B., Somers, T. J., Keefe, F. J., Gen. Hosp. Psychiatry 31, 206–219 (2009).
1195–1201 (2007). Mosley-Williams, A. & Lumley, M. A. Pain and 101. Doherty, M. et al. A randomised controlled trial of
66. Wolfe, F. & Michaud, K. Predicting depression in functioning of rheumatoid arthritis patients ibuprofen, paracetamol or a combination tablet
rheumatoid arthritis: the signal importance of based on marital status: is a distressed of ibuprofen/paracetamol in community-derived
pain extent and fatigue, and comorbidity. Arthritis marriage preferable to no marriage? J. Pain 11, people with knee pain. Ann. Rheum. Dis. 70,
Rheum. 61, 667–673 (2009). 958–964 (2012). 1534–1541 (2011).
67. Fifield, J., Tennen, H., Reisine, S. & McQuillan, J. 84. Waltz, M., Kriegel, W. & van’t Pad Bosch, P. 102. Zhang, W., Robertson, J., Jones, A. C.,
Depression and the long-term risk of pain, The social environment and health in rheumatoid Dieppe, P. A. & Doherty, M. The placebo effect
fatigue, and disability in patients with arthritis: marital quality predicts individual and its determinants in osteoarthritis: meta-
rheumatoid arthritis. Arthritis Rheum. 41, variability in pain severity. Arthritis Care Res. analysis of randomised controlled trials.
1851–1857 (1998). (Hoboken) 11, 356–374 (1998). Ann. Rheum. Dis. 67, 1716–1723 (2008).
68. Zautra, A. J. et al. Depression history, stress, 85. Mapp, P. I. & Walsh, D. A. Mechanisms and 103. Frank, R. G. et al. Antidepressant analgesia
and pain in rheumatoid arthritis patients. targets of angiogenesis and nerve growth in in rheumatoid arthritis. J. Rheumatol. 15,
J. Behav. Med. 30, 187–197 (2007). osteoarthritis. Nat. Rev. Rheumatol. 8, 390–398 1632–1638 (1988).
69. Conner, T. S. et al. Coping with rheumatoid (2012). 104. Lemmel, E. M. et al. Efficacy and safety of
arthritis pain in daily life: within-person analyses 86. Black, P. H. Stress and the inflammatory meloxicam in patients with rheumatoid arthritis.
reveal hidden vulnerability for the formerly response: a review of neurogenic inflammation. J. Rheumatol. 24, 282–290 (1997).
depressed. Pain 126, 198–209 (2006). Brain Behav. Immun. 16, 622–653 (2002). 105. Blyth, T., Hunter, J. A. & Stirling, A. Pain relief
70. Rupp, I., Boshuizen, H. C., Dinant, H. J., 87. Edwards, R. R. et al. Association of in the rheumatoid knee after steroid injection.
Jacobi, C. E. & van den Bos, G. A. M. Disability catastrophizing with interleukin-6 responses to A single-blind comparison of hydrocortisone
and health-related quality of life among patients acute pain. Pain 140, 135–144 (2008). succinate, and triamcinolone acetonide or
with rheumatoid arthritis: association with 88. Geenen, R., van Middendorp, H. & Bijlsma, J. W. hexacetonide. Br. J. Rheumatol. 33, 461–463
radiographic joint damage, disease activity, pain, The impact of stressors on health status and (1994).
and depressive symptoms. Scand. J. Rheumatol. hypothalamic–pituitary–adrenal axis and 106. Kirwan, J. R. et al. A randomised placebo
35, 175–181 (2006). autonomic nervous system responsiveness in controlled 12 week trial of budesonide and
71. Smedstad, L. M., Vaglum, P., Kvien, T. K. & rheumatoid arthritis. Ann. NY Acad. Sci. 1069, prednisolone in rheumatoid arthritis.
Moum, T. The relationship between self-reported 77–97 (2006). Ann. Rheum. Dis. 63, 688–695 (2004).
pain and sociodemographic variables, anxiety, 89. Dekkers, J. C. et al. Biopsychosocial mediators 107. Choy, E. H., Kingsley, G. H., Corkill, M. M. &
and depressive symptoms in rheumatoid and moderators of stress-health relationships in Panayi, G. S. Intramuscular methylprednisolone
arthritis. J. Rheumatol. 22, 514–520 (1995). patients with recently diagnosed rheumatoid is superior to pulse oral methylprednisolone
72. Keefe, F. J. et al. Pain coping strategies and arthritis. Arthritis Rheum. 45, 307–316 (2001). during the induction phase of chrysotherapy.
coping efficacy in rheumatoid arthritis: a daily 90. Steptoe, A., Hamer, M. & Chida, Y. The effects of Br. J. Rheumatol. 32, 734–739 (1993).
process analysis. Pain 69, 35–42 (1997). acute psychological stress on circulating 108. Kirwan, J. R. The effect of glucocorticoids on
73. Urrows, S., Affleck, G., Tennen, H. & Higgins, P. inflammatory factors in humans: a review and joint destruction in rheumatoid arthritis. The
Unique clinical and psychological correlates of meta-analysis. Brain Behav. Immun. 21, Arthritis and Rheumatism Council Low-Dose
fibromyalgia tender points and joint tenderness 901–912 (2007). Glucocorticoid Study Group. N. Engl. J. Med.
in rheumatoid arthritis. Arthritis Rheum. 37, 91. Pace, T. W. & Heim, C. M. A short review on the 333, 142–146 (1995).
1513–1520 (1994). psychoneuroimmunology of posttraumatic 109. Hickling, P., Jacoby, R. K. & Kirwan, J. R. Joint
74. Lee, Y. C. et al. The relationship between disease stress disorder: from risk factors to medical destruction after glucocorticoids are withdrawn
activity, sleep, psychiatric distress and pain comorbidities. Brain Behav. Immun. 25, 6–13 in early rheumatoid arthritis. Arthritis and
sensitivity in rheumatoid arthritis: a cross- (2011). Rheumatism Council Low Dose Glucocorticoid
sectional study. Arthritis Res. Ther. 11, R160 92. Gimeno, D. et al. Adult socioeconomic position, Study Group. Br. J. Rheumatol. 37, 930–936
(2009). C-reactive protein and interleukin-6 in the (1998).
75. Dharmshaktu, P., Tayal, V. & Kalra, B. S. Efficacy Whitehall II prospective study. Eur. J. Epidemiol. 110. Williams, H. J. et al. Comparison of low-dose
of antidepressants as analgesics: a review. 22, 675–683 (2007). oral pulse methotrexate and placebo in the
J. Clin. Pharmacol. 52, 6–17 (2012). 93. Qureshi, S. U., Pyne, J. M., Magruder, K. M., treatment of rheumatoid arthritis. A controlled
76. Zautra, A. J. et al. Comparison of cognitive Schulz, P. E. & Kunik, M. E. The link between clinical trial. Arthritis Rheum. 28, 721–730
behavioral and mindfulness meditation post-traumatic stress disorder and physical (1985).
interventions on adaptation to rheumatoid comorbidities: a systematic review. Psychiatr. Q. 111. Pinals, R. S., Kaplan, S. B., Lawson, J. G. &
arthritis for patients with and without history of 80, 87–97 (2009). Hepburn, B. Sulfasalazine in rheumatoid
recurrent depression. J. Consult. Clin. Psychol. 94. Peters, M. J. et al. Genome-wide association arthritis. A double-blind, placebo-controlled trial.
76, 408–421 (2008). study meta-analysis of chronic widespread pain: Arthritis Rheum. 29, 1427–1434 (1986).

590  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

112. Smolen, J. S. et al. Efficacy and safety of 126. Allaart, C. F. & Huizinga, T. W. Treatment behaviour in monoarthritic rats. Pain 116,
leflunomide compared with placebo and strategies in recent onset rheumatoid arthritis. 411–419 (2005).
sulphasalazine in active rheumatoid arthritis: Curr. Opin. Rheumatol. 23, 241–244 (2011). 145. Sharif Naeini, R., Cahill, C. M., Ribeiro-da-
a double-blind, randomised, multicentre trial. 127. Schaible, H. G. et al. The role of proinflammatory Silva, A., Menard, H. A. & Henry, J. L.
European Leflunomide Study Group. Lancet 353, cytokines in the generation and maintenance of Remodelling of spinal nociceptive mechanisms
259–266 (1999). joint pain. Ann. NY Acad. Sci. 1193, 60–69 in an animal model of monoarthritis. Eur. J.
113. National Collaborating Centre for Chronic (2010). Neurosci. 22, 2005–2015 (2005).
conditions (UK). Rheumatoid arthritis: national 128. Schipper, L. G., Fransen, J., den Broeder, A. A. 146. Abbadie, C., Besson, J. M. & Calvino, B. c-Fos
clinical guidelines for management and & Van Riel, P. L. Time to achieve remission expression in the spinal cord and pain-related
treatment in adults (NICE Clinical Guidelines, determines time to be in remission. Arthritis symptoms induced by chronic arthritis in the rat
No. 79) [online], https://www.ncbi.nlm.nih.gov/ Res. Ther. 12, R97 (2010). are prevented by pretreatment with Freund
books/NBK51812/ (2009). 129. Pradhan, E. K. et al. Effect of mindfulness-based adjuvant. J. Neurosci. 14, 5865–5871 (1994).
114. van der Kooij, S. M. et al. Patient-reported stress reduction in rheumatoid arthritis 147. Chen, Y., Willcockson, H. H. & Valtschanoff, J. G.
outcomes in a randomized trial comparing four patients. Arthritis Rheum. 57, 1134–1142 Influence of the vanilloid receptor TRPV1 on the
different treatment strategies in recent-onset (2007). activation of spinal cord glia in mouse models of
rheumatoid arthritis. Arthritis Rheum. 61, 4–12 130. Knittle, K., Maes, S. & de Gucht, V. Psychological pain. Exp. Neurol. 220, 383–390 (2009).
(2009). interventions for rheumatoid arthritis: examining 148. Hains, L. E. et al. Pain intensity and duration can
115. Weinblatt, M. E. et al. A trial of etanercept, a the role of self-regulation with a systematic be enhanced by prior challenge: initial evidence
recombinant tumor necrosis factor receptor:Fc review and meta-analysis of randomized suggestive of a role of microglial priming. J. Pain
fusion protein, in patients with rheumatoid controlled trials. Arthritis Care Res. (Hoboken) 11, 1004–1014 (2010).
arthritis receiving methotrexate. N. Engl. J. Med. 62, 1460–1472 (2010). 149. Bao, L. et al. Adjuvant-induced arthritis: IL-1 β,
340, 253–259 (1999). 131. Koltyn, K. F. & Umeda, M. Exercise, hypoalgesia IL-6 and TNF-α are up-regulated in the spinal
116. Keystone, E. C. et al. Radiographic, clinical, and blood pressure. Sports Med. 36, 207–214 cord. Neuroreport 12, 3905–3908 (2001).
and functional outcomes of treatment with (2006). 150. Honor, P. et al. Spinal substance P receptor
adalimumab (a human anti-tumor necrosis factor 132. Tendzegolskis, Z., Viru, A. & Orlova, E. Exercise- expression and internalization in acute, short-
monoclonal antibody) in patients with active induced changes of endorphin contents in term, and long-term inflammatory pain states.
rheumatoid arthritis receiving concomitant hypothalamus, hypophysis, adrenals and blood J. Neurosci. 19, 7670–7678 (1999).
methotrexate therapy: a randomized, placebo- plasma. Int. J. Sports Med. 12, 495–497 151. Pezet, S., Onteniente, B., Grannec, G.
controlled, 52-week trial. Arthritis Rheum. 50, (1991). & Calvino, B. Chronic pain is associated
1400–1411 (2004). 133. Guieu, R., Blin, O., Pouget, J. & Serratrice, G. with increased TrkA immunoreactivity in
117. Andratsch, M. et al. A key role for gp130 Nociceptive threshold and physical activity. spinoreticular neurons. J. Neurosci. 19,
expressed on peripheral sensory nerves in Can. J. Neurol. Sci. 19, 69–71 (1992). 5482–5492 (1999).
pathological pain. J. Neurosci. 29, 134. Baillet, A. et al. Efficacy of cardiorespiratory 152. Sun, S. et al. New evidence for the involvement
13473–13483 (2009). aerobic exercise in rheumatoid arthritis: meta- of spinal fractalkine receptor in pain facilitation
118. Russell, F. A., Fernandes, E. S., Courade, J. P., analysis of randomized controlled trials. Arthritis and spinal glial activation in rat model of
Keeble, J. E. & Brain, S. D. Tumour necrosis Care Res. (Hoboken) 62, 984–992 (2010). monoarthritis. Pain 129, 64–75 (2007).
factor α mediates transient receptor potential 135. Uhlig, T., Fongen, C., Steen, E., Christie, A. & 153. Guilbaud, G., Benoist, J. M., Gautron, M. &
vanilloid 1-dependent bilateral thermal Odegard, S. Exploring Tai Chi in rheumatoid Kayser, V. Aspirin clearly depresses responses of
hyperalgesia with distinct peripheral roles arthritis: a quantitative and qualitative study. ventrobasal thalamus neurons to joint stimuli in
of interleukin-1β, protein kinase C and BMC Musculoskelet. Disord. 11, 43 (2010). arthritic rats. Pain 13, 153–163 (1982).
cyclooxygenase-2 signalling. Pain 142, 264–274 136. Kosek, E., Ekholm, J. & Hansson, P. Modulation 154. Neto, F. L. et al. Supraspinal metabolic activity
(2009). of pressure pain thresholds during and following changes in the rat during adjuvant
119. Nishimoto, N. et al. Study of active controlled isometric contraction in patients with monoarthritis. Neuroscience 94, 607–621
monotherapy used for rheumatoid arthritis, fibromyalgia and in healthy controls. Pain 64, (1999).
an IL-6 inhibitor (SAMURAI): evidence of clinical 415–423 (1996). 155. Omura, K., Imai, S., Maeda, T. & Hukuda, S.
and radiographic benefit from an X ray reader- 137. Radboud University Nijmegen Medical Center. Prolonged and increasing expression of Fos
blinded randomised controlled trial of DAS-score.nl: disease activity score in rheumatoid related antigens in the hippocampus of adjuvant
tocilizumab. Ann. Rheum. Dis. 66, 1162–1167 arthritis [online], http://www.reuma-nijmegen.nl/ arthritic rats. J. Rheumatol. 25, 936–944
(2007). www.das-score.nl/index.html (2014). (1998).
120. Rech, J. et al. Association of brain functional 138. Janta, I. et al. Patient self-assessment and 156. Williams, F. G. & Beitz, A. J. Chronic pain
magnetic resonance activity with response to physician’s assessment of rheumatoid arthritis increases brainstem proneurotensin/
tumor necrosis factor inhibition in rheumatoid activity: which is more realistic in remission neuromedin-N mRNA expression:
arthritis. Arthritis Rheum. 65, 325–333 status? A comparison with ultrasonography. a hybridization-histochemical and
(2013). Rheumatology (Oxford) http://dx.doi.org/ immunohistochemical study using three
121. Drossaers-Bakker, K. W. et al. Long-term 10.1093/rheumatology/ket297 (2013). different rat models for chronic nociception.
outcome in rheumatoid arthritis: a simple 139. Wakefield, R. J. et al. Should oligoarthritis be Brain Res. 611, 87–102 (1993).
algorithm of baseline parameters can predict reclassified? Ultrasound reveals a high 157. Xie, H., Ma, F., Zhang, Y.-Q., Gao, X. & Wu, G.-C.
radiographic damage, disability, and disease prevalence of subclinical disease. Ann. Rheum. Expression of 5-HT2A receptor mRNA in some
course at 12-year followup. Arthritis Rheum. 47, Dis. 63, 382–385 (2004). nuclei of brain stem enhanced in monoarthritic
383–390 (2002). 140. Shinoda, M. et al. Nerve terminals extend into rats. Brain Res. 954, 94–99 (2002).
122. Sokka, T., Kankainen, A. & Hannonen, P. the temporomandibular joint of adjuvant arthritic 158. Danziger, N., Weil-Fugazza, J., Le Bars, D.
Scores for functional disability in patients with rats. Eur. J. Pain 7, 493–505 (2003). & Bouhassira, D. Alteration of descending
rheumatoid arthritis are correlated at higher 141. Yamazaki, Y., Ren, K., Shimada, M. & Iwata, K. modulation of nociception during the course
levels with pain scores than with radiographic Modulation of paratrigeminal nociceptive of monoarthritis in the rat. J. Neurosci. 19,
scores. Arthritis Rheum. 43, 386–389 (2000). neurons following temporomandibular joint 2394–2400 (1999).
123. Sarzi-Puttini, P. et al. Correlation of the score for inflammation in rats. Exp. Neurol. 214, 209–218 159. Constandil, L. et al. Effect of interleukin-1β on
subjective pain with physical disability, clinical (2008). spinal cord nociceptive transmission of normal
and radiographic scores in recent onset 142. Schaible, H. G., Ebersberger, A. & Von and monoarthritic rats after disruption of glial
rheumatoid arthritis. BMC Musculoskelet. Disord. Banchet, G. S. Mechanisms of pain in arthritis. function. Arthritis Res. Ther. 11, R105 (2009).
3, 18 (2002). Ann. NY Acad. Sci. 966, 343–354 (2002). 160. Clark, A. K., Grist, J., Al-Kashi, A., Perretti, M. &
124. Judge, A. et al. Predictors of outcomes of total 143. Imbe, H. et al. Orofacial deep and cutaneous Malcangio, M. Spinal cathepsin S and fractalkine
knee replacement surgery. Rheumatology tissue inflammation and trigeminal neuronal contribute to chronic pain in the collagen-
(Oxford) 51, 1804–1813 (2012). activation. Implications for persistent induced arthritis model. Arthritis Rheum. 64,
125. Dore, D. et al. Natural history and clinical temporomandibular pain. Cells Tissues Organs 2038–2047 (2012).
significance of MRI-detected bone marrow 169, 238–247 (2001). 161. Christianson, C. A. et al. Characterization of
lesions at the knee: a prospective study in 144. Cruz, C. D., Neto, F. L., Castro-Lopes, J., the acute and persistent pain state present in
community dwelling older adults. Arthritis Res. McMahon, S. B. & Cruz, F. Inhibition of ERK K/BxN serum transfer arthritis. Pain 151,
Ther. 12, R223 (2010). phosphorylation decreases nociceptive 394–403 (2010).

NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  OCTOBER 2014  |  591


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

162. von Banchet, G. S., Petrow, P. K., Brauer, R. & 166. Natura, G. et al. Neuronal prostaglandin E2 171. Whittle, S. L., Richards, B. L., van der
Schaible, H. G. Monoarticular antigen-induced receptor subtype EP3 mediates antinociception Heijde, D. M. & Buchbinder, R. The efficacy
arthritis leads to pronounced bilateral during inflammation. Proc. Natl Acad. Sci. USA and safety of opioids in inflammatory arthritis:
upregulation of the expression of neurokinin 1 110, 13648–13653 (2013). a Cochrane systematic review. J. Rheumatol.
and bradykinin 2 receptors in dorsal root ganglion 167. Hazlewood, G., van der Heijde, D. M. & Suppl. 90, 40–46 (2012).
neurons of rats. Arthritis Res. 2, 424–427 (2000). Bombardier, C. Paracetamol for the management 172. Richards, B. L., Whittle, S. L., van der
163. Segond von Banchet, G., Petrow, P. K., Brauer, R. of pain in inflammatory arthritis: a systematic Heijde, D. M. & Buchbinder, R. The efficacy
& Schaible, H. G. Monoarticular antigen-induced literature review. J. Rheumatol. Suppl. 90, 11–16 and safety of antidepressants in inflammatory
arthritis leads to pronounced bilateral (2012). arthritis: a Cochrane systematic review.
upregulation of the expression of neurokinin 1 168. Wienecke, T. & Gotzsche, P. C. Paracetamol J. Rheumatol. Suppl. 90, 21–27 (2012).
and bradykinin 2 receptors in dorsal root versus nonsteroidal anti-inflammatory drugs for 173. Richards, B. L., Whittle, S. L. & Buchbinder, R.
ganglion neurons of rats. Arthritis Res. 2, rheumatoid arthritis. Cochrane Database of Neuromodulators for pain management in
424–427 (2000). Systematic Reviews 2004, Issue 1. Art. No.: rheumatoid arthritis. Cochrane Database of
164. Boettger, M. K., Weber, K., Gajda, M., Brauer, R. CD003789. http://dx.doi.org/10.1002/ Systematic Reviews 2012, Issue 1. Art. No.:
& Schaible, H. G. Spinally applied ketamine or 14651858.CD003789.pub2. CD008921. http://dx.doi.org/10.1002/
morphine attenuate peripheral inflammation and 169. McCormack, P. L. Celecoxib: a review of its use 14651858.CD008921.pub2.
hyperalgesia in acute and chronic phases of for symptomatic relief in the treatment of 174. International Association for the Study of Pain.
experimental arthritis. Brain Behav. Immun. 24, osteoarthritis, rheumatoid arthritis and IASP Taxonomy: pain terms [online], http://
474–485 (2010). ankylosing spondylitis. Drugs 71, 2457–2489 www.iasp-pain.org/Education/Content.
165. Boettger, M. K. et al. Spinal tumor necrosis (2011). aspx?ItemNumber=1698 (2012).
factor α neutralization reduces peripheral 170. Chen, Y. F. et al. Cyclooxygenase-2 selective
inflammation and hyperalgesia and suppresses non-steroidal anti-inflammatory drugs (etodolac,
autonomic responses in experimental arthritis: meloxicam, celecoxib, rofecoxib, etoricoxib, Author contributions
a role for spinal tumor necrosis factor α during valdecoxib and lumiracoxib) for osteoarthritis Both authors researched the data for the article,
induction and maintenance of peripheral and rheumatoid arthritis: a systematic review provided substantial contributions to discussions
inflammation. Arthritis Rheum. 62, 1308–1318 and economic evaluation. Health Technol. of content, wrote the article and undertook review
(2010). Assess. 12, 1–278 (2008). and/or editing of the manuscript before submission.

592  |  OCTOBER 2014  |  VOLUME 10  www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved

Вам также может понравиться