Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: J Pain. 2013 Dec;14(12):1539–1552. doi: 10.1016/j.jpain.2013.08.007

The association of sleep and pain: An update and a path forward

Patrick H Finan 1,*, Burel R Goodin 2, Michael T Smith 1
PMCID: PMC4046588  NIHMSID: NIHMS521705  PMID: 24290442

Abstract

Ample evidence suggests that sleep and pain are related. However, many questions remain about the direction of causality in their association, as well as mechanisms that may account for their association. The prevailing view has generally been that they are reciprocally related. The present review critically examines the recent prospective and experimental literature (2005-present) in an attempt to update the field on emergent themes pertaining to the directionality and mechanisms of the association of sleep and pain. A key trend emerging from population-based longitudinal studies is that sleep impairments reliably predict new incidents and exacerbations of chronic pain. Microlongitudinal studies employing deep subjective and objective assessments of pain and sleep support the notion that sleep impairments are a stronger, more reliable predictor of pain than pain is of sleep impairments. Recent experimental studies suggest that sleep disturbance may impair key processes that contribute to the development and maintenance of chronic pain, including endogenous pain inhibition and joint pain. Several biopsychosocial targets for future mechanistic research on sleep and pain are discussed, including dopamine and opioid systems, positive and negative affect, and sociodemographic factors.

Perspective

This critical review examines the recent prospective and experimental research (2005-present) on the association of sleep and pain in an attempt to identify trends suggestive of directionality and potential mechanisms. An update on this literature is needed to guide future clinical efforts to develop and augment treatments for chronic sleep disturbance and chronic pain.

Keywords: Chronic pain, sleep, insomnia, longitudinal, sleep deprivation

Introduction

Pain is a physical and emotional signal of bodily harm that strongly motivates behavior.. Sleep is a behaviorally regulated drive that broadly serves to maintain homeostasis and optimize function across multiple physiologic systems. Humans require both pain and sleep for survival; however, chronic impairments in the systems regulating pain and sleep can have a broad negative impact on health and well-being. Sleep complaints are present in 67-88% of chronic pain disorders 70,103 and at least 50% of individuals with insomnia—the most commonly diagnosed disorder of sleep impairment—suffer from chronic pain 117. Across most medical interventions, the development of pain as a side effect coincides with the development of sleep disturbance, and vice-versa 20. Further, both chronic pain and sleep disturbances share an array of physical and mental health comorbidities, such as obesity 45, type 2 diabetes 18,50, and depression 34,127.

In the backdrop of this accruing evidence in support of a sleep/pain association, two fundamental questions continue to linger: 1) Are pain and sleep reciprocally or unidirectionally related? and 2) What mechanisms account for the associations between sleep and pain? The vast expanse of research on the association of sleep and pain necessitates that we narrow the scope of the present review. Prior reviews on the subject have examined evidence from prospective 103 and experimental 54 studies to determine if sleep and pain are reciprocally related or better characterized through unidirectional models. The steadily emergent view supported by those reviews has been that pain and sleep are reciprocally related, and that acute experimentally-induced sleep loss increases pain sensitivity. However, early evidence informing those views was limited by methodological inconsistencies across a relatively small number of studies, and newer data suggest a more complex and textured characterization is warranted. As methods of prospective and experimental design have advanced in recent years, our intention is to provide an updated review of prospective and experimental studies conducted in the past decade to clarify what we currently know about the issue of reciprocality/directionality. Additionally, we discuss several promising targets for future research on potential mechanisms underlying the association of sleep and pain. Although we have not exhausted the list of possible mechanisms, we focus on three mechanistic possibilites (affective systems, brain neurotransmitter systems, and sociodemographic factors) that have received considerable attention in the separate fields of sleep and pain research, but have received less integrative attention as mechanisms of the sleep/pain interactions The pharmacological 6 and cognitive-behavioral 116 treatment outcome literatures have been recently reviewed elsewhere. Therefore, we will not review these studies, and refer readers to those manuscripts.

Methods

Smith and Haythornthwaite 103 discussed longitudinal studies published in the years prior to and including 2004. Therefore, we searched the PubMed and Google Scholar databases for longitudinal studies, restricting the search to studies published from 2005-present, and employing the following search terms individually and in combination: “pain,” “chronic pain,” “sleep,” “insomnia,” “longitudinal,” “prospective,” and “daily diary.” Lautenbacher et al. 54 discussed experimental studies published in the years prior to and including 2006. Therefore, our search for experimental studies was conducted in PubMed and Google Scholar, restricting the search to studies published from 2006-present, and employing the following search terms individually and in combination: “pain,” “pain sensitivity,” “hyperalgesia,” “quantitative sensory testing,” “sleep deprivation,” “total sleep deprivation,” “partial sleep deprivation,” “sleep fragmentation,” and “experimental.”

In addition to those database searches, reference sections of relevant studies were scanned for additional articles not identified by the original search. All prospective and experimental sleep deprivation studies identified were included in this review. Further, any additional article appearing in the synthesis of the prospective and experimental studies was chosen for review based on its relevance, in our judgment, for addressing the questions of directionality and mechanisms of the association of sleep and pain. Articles were neither included nor excluded based on the statistical significance, or lack thereof, of the results.

We first review the key findings from studies identified in our search procedure. Subsequently, we raise several directions for future research, organized under two broad headings: 1) Biobehavioral mechanisms of the association of sleep and pain; and 2) Sociodemographic moderators of the association of sleep and pain. Although these categories by no means exhaust the potential supply of future studies to enhance our understanding of sleep and pain, we have selected them because they represent large gaps in the current knowledge base and may be incorporated into existing programs of research with relative ease.

Results

Directionality or Reciprocal Influence?

Overview of Prior Work

Early longitudinal evidence reviewed by Smith & Haythornthwaite (2) suggested a reciprocal relationship between sleep and pain. In that review, a pain→sleep directional effect was observed in 5/6 applicable longitudinal studies 1,21,73,93,94, and evidence for a sleep→pain directional effect in 4/6 applicable studies 1,21,93,112 involving patients with fibromyalgia, rheumatoid arthritis, burn injury, and orofacial pain. Since then, the literature has matured in several key facets that improve the strength of inferences that can be made. First, there are over twice as many prospective studies (see: Table 1) that include a variety of novel clinical samples, including patients with tension headache, migraine, primary insomnia, primary depression, and pediatric chronic pain. Second, there is now a greater mix of longitudinal (e.g., few time points, far apart) and microlongitudinal (e.g., many time points, close together) studies, providing greater texture in the temporal analysis of sleep and pain. Third, several studies examine new instances of pain complaints and/or sleep disturbance predicted by prior symptoms. Fourth, there are now several very large epidemiological studies from which one may more firmly draw population-level conclusions.

Table 1. Longitudinal and Microlongitudinal Studies 2005-2012.
Reference Study Sample Pain Measures Sleep Measures Pain→Sleep Sleep→Pain
Lyngberg et al. 62 Tension Headache
N = 549
and Migraine
N = 160
Chronic headache (12-year follow-up) Self-report sleep problems (baseline) N/A Yes*
Boardman et al. 10 Headache-free general population
N = 455
Headache incidence (1-year follow-up) Self-report severe sleep problems (baseline) N/A Yes
Hamilton et al. 43 Fibromyalgia and rheumatoid arthritis
N = 49
  1. Correlation of daily pain and negative affect

  2. Correlation of daily pain and positive affect

  1. Sleep quality

  2. Sleep duration (both baseline)

N/A
  1. Yes**

  2. Yes*

Davies et al. 17 Chronic widespread pain
N = 679
Resolution of chronic widespread pain (15 months follow-up) Self-report restorative sleep (baseline) N/A Yes*
Bigatti et al. 8a Fibromyalgia
N = 492
Self-report pain (baseline; 1 year) Self-report sleep quality (baseline; 1 year) N/S Yes*
Edwards et al. 29 General population
N = 971
Daily self-report pain Daily self-report sleep duration Yes** (weaker) Yes** (stronger)
Smith et al. 105 Burn injury
N = 333
Self-report pain (baseline; 2 year) Self-report sleep-onset insomnia (baseline; 2 year) Yes** Yes**
Quartana et al. 88 TMD
N = 53
Bi-weekly pain ratings Bi-weekly insomnia severity N/S Yes*
Dzierzewski et al. 24 Older adults with insomnia
N = 50
Daily self-report pain Actigraphy N/A Yes*
Chung & Tso 16 Major Depression
N = 82
Self-report pain (baseline; 3 month) Actigraphy (baseline; 3 month) N/A Yes*
O'Brien et al. 76 Fibromyalgia
N = 22
Daily self-report pain
  1. Daily self-report sleep quality

  2. Actigraphy

  1. Yes**

  2. N/S

  1. Yes**

  2. N/S

Lewandowski et al. 59 Adolescents w/Chronic Pain
N = 39
Daily self-report pain Actigraphy N/S Yes**
Odegard et al. 77 General population
N = 15,268
Headache incidence (11 year follow-up) Self-report insomnia (baseline) N/A Yes**
Bromberg et al. 12 Adolescents w/Juvenile Polyarticular Arthritis
N = 51
Daily self-report pain Daily self-report sleep quality N/S Yes**
Mork & Nilsen 71 General population
N = 12,350
Fibromyalgia incidence (11 year follow-up) Self-report insomnia (baseline) N/A Yes**
Jansson-Frojmark & Boersma 47 General population
N = 1,746
  1. Pain incidence

  2. Pain persistence (baseline; 1 year)

  1. Insomnia incidence

  2. Insomnia persistence (baseline; 1 year)

  1. Yes*

  2. Yes*

  1. N/S

  2. Yes**

Tang et al. 115 Comorbid heterogeneous chronic pain & insomnia
N = 119
Daily self-report pain Daily self-report sleep quality, efficiency Yes* (weaker) Yes* (stronger)

Note. N/A = not assessed; N/S = not significant; ‘stronger’ and ‘weaker’ refer to the relative strength of the general directional effect across multiple data analytic models tested.

a

This study tested various sequential models with path analysis, and only reported the statistics for the best fitting model, in which sleep quality predicted pain.

*

p < .05

**

p < .01

p value not reported

We broadly categorized the recent prospective studies into those that only assessed the sleep→pain directional effect and those that assessed a bidirectional association. We did not find any recent prospective studies that have exclusively evaluated the unidirectional pain→sleep directional effect.

Recent Prospective Studies Assessing the Unidirectional Effect of Sleep on Future Pain (2005-2012)

Three large longitudinal studies have demonstrated that elevated insomnia symptoms increase the risk of exacerbating existing headache and developing new incident headache at long-term follow-up, ranging from 1-12 years 10,62,77. Specifically, Danish individuals with infrequent, episodic tension-type headache were more likely to develop chronic tension-type headache at 12-year follow-up if insomnia symptoms were present at baseline 62. Insomnia symptoms, however, were not predictive of migraine prognosis in that sample. In contrast, baseline insomnia symptoms in a headache-free, population-based Norwegian sample predicted new incident cases of both tension-type and migraine headache at an 11-year follow-up 77. A headache-free, population-based British sample were significantly more likely to develop new incident cases of headache (diagnostic type not specified) at 1-year follow-up if insomnia symptoms were present at baseline 10. In the same study, individuals with headache were more likely to remit at 1 year if insomnia symptoms were absent at baseline 10.

A large population-based study of Norwegian women found that women who endorse frequent, “sleep problems,” defined as frequent difficulty falling asleep or having a sleep disorder, were significantly more likely to develop fibromyalgia 10 years later 71. The authors estimated that 2/3 of the incident cases of fibromyalgia in their sample were explained by sleep problems 71. These findings are supported by a separate population-based study that found that insomnia symptoms at baseline significantly increased the risk of developing chronic musculoskeletal pain (both widespread and regional) at 17-year follow-up 74. Quality sleep has also been shown to predict chronic widespread pain symptom resolution over 15 months 17.

In addition to the longitudinal studies, recent microlongitidinal studies have shown sleep disruption to linearly predict next-day pain reports in patients with depression 16 and older adults 24, and next-day affective responses to pain in RA and fibromyalgia patients 43.

Together, these prospective studies indicate that sleep disturbance increases the risk for new-onset cases of chronic pain in pain free individuals, worsens the long-term prognosis of existing headache and chronic musculoskeletal pain, and influences daily fluctuations in clinical pain. Furthermore, good sleep appears to improve the long-term prognosis of individuals with tension-type headache, migraine, and chronic musculoskeletal pain.

Recent Prospective Studies Assessing Bidirectional Effects of Sleep and Pain (2005-2012)

Evidence for Temporal Precedence of Sleep Over Pain

Across the studies investigating bidirectional linkages, a trend has emerged suggesting that sleep disturbance may be a stronger predictor of pain than pain of sleep disturbance. A 1-year longitudinal cohort study of fibromyalgia patients revealed through structural equation modeling that sleep disturbance temporally preceded increases in pain, whereas pain at Time 1 was not significantly associated with sleep disturbance 1 year later 8. Cross-lagged panel modeling of bi-weekly insomnia severity and pain ratings in temporomandibular disorder (TMD) patients found a similar effect 88. Across 3 months of measurement (6 bi-weekly assessments) within-month fluctuations in insomnia severity predicted next-month changes in pain ratings, whereas fluctuations in pain were not predictive of future changes in insomnia severity. In the general population, sleep and pain evidenced lagged day-to-day interdependencies 29. However, the magnitude of the effect was stronger for the sleep→pain direction than for the opposite direction, such that decreased sleep on a given day predicted increased pain on the subsequent day 29.

Evidence favoring the temporal precedence of sleep over pain was also presented in 3 subsequent studies of daily sleep and pain. In one study of adolescents with heterogeneous chronic pain complaints, including headache, abdominal pain, back pain, other musculoskeletal pain, daily actigraphy assessments revealed significant associations of total sleep time and wake after sleep onset on next-day pain reports 59. Pain did not prospectively predict any sleep measure. In another study of adolescents with juvenile polyarticular arthritis, daily self-reported poor sleep quality predicted daily pain, but the reverse association was not significant 12.Similar effects were observed in adult pain clinic patients with a wide range of chronic pain etiologies and comorbid insomnia 115. In that study, self-reported sleep quality and actigraphically-measured sleep efficiency reliably predicted next-day pain reports. In contrast, pain was removed from final models predicting sleep parameters due to its relatively weak predictive validity compared to cognitive variables.

It is worth noting that, in addition to these prospective studies, a large cross-sectional cohort study of cancer patients utilized structural equation modeling to suggest directionality in the sleep/pain association 111. In a series of analyses involving associations between depression, fatigue, sleep, and pain, the best fitting structural equation model included a path in which sleep predicted pain; inclusion of the reverse path—pain predicting sleep—produced a poorer fitting model that was dropped from final analyses. These findings provide suggestive evidence that the influence of sleep on pain symptoms may be stronger than the influence of pain on sleep symptoms in cancer patients.

Evidence for Bidirectionality

Other recent longitudinal studies have offered data consistent with a bidirectional association of sleep and pain. A large longitudinal study of acute burn injury patients employed linear mixed modeling and demonstrated that acute insomnia symptoms observed at hospital discharge predicted attenuated recovery and elevated pain severity over a 2-year follow-up period 105. The reverse relationship was also observed: pain severity at discharge predicted insomnia severity at 2-year follow-up. A reciprocal relationship between chronic pain and chronic insomnia symptoms was observed over a 1 year time frame in a population-based study of Swedish adults 47. However, insomnia at baseline did not significantly predict new incidents of pain at follow-up after adjusting for age, sex, and depression symptoms. On a daily time scale, a bidrectional association of pain and sleep disturbance was observed in a sample of women with fibromyalgia, 76.In that study, multilevel modeling revealed that poor self-reported subjective sleep quality predicted increased next-day pain, and increased pain predicted poor next-night sleep quality.

Summary of Recent Prospective Studies

As experimental and data analytic methods have been refined over the past decade, a trend in the literature suggests that the temporal effect of sleep on pain may be stronger than that of pain on sleep. Out of 9 recent prospective studies that have tested both directional effects, 6 found stronger evidence for the temporal precedence of sleep over pain 8,12,29,59,88,115. Each of these studies employed sophisticated measurement (e.g., microlongitudinal) and analytic techniques, including multilevel modeling and cross-lagged panel modeling, whereby time-based dependencies in repeated measures and random error variance are controlled, and sequential relationships can be inferred with a greater degree of confidence than is possible with ordinary least squares regression or ANOVA techniques. Notably, 2 of the studies that reported relatively equivalent bidirectional effects 47,105 were limited by their reliance on basic self-report instruments to assess sleep and pain. Thus, when assessed in broad strokes, sleep and pain may appear to be reciprocally related, whereas finer-grained analyses suggest that poor sleep may exert a stronger and perhaps more durable toll on the experience of chronic pain (see O'Brien et al. 76 as a notable exception). Further, several large prospective studies suggest that sleep problems increase the risk of developing future chronic pain, and good sleep increases the chance that chronic pain will remit over time 10,17,62,71,74,77. These studies provide a sound basis from which to hypothesize that sleep has a causal influence on pain. From a clinical standpoint, these findings strongly suggest that sleep disruption may hold significant promise as an intervention target in efforts to prevent and treat chronic pain. However, it is important to note that it is not clear if different mechanisms are at play in the development of new incident pain as opposed to exacerbation of existing pain as a result of sleep disturbance. Future work should investigate if the mechanisms are identical, or if the sleep disturbance input required to provoke a new incident case is of greater magnitude or duration than that required to provoke a flare of an existing condition.

Going forward, prospective studies should also include more objective measures of sleep and pain. Several studies reviewed here employed actigraphy, which provides an objective assessment of activity versus inactivity to quantify sleep parameters, including total sleep time, sleep onset latency, wake after sleep onset, and sleep efficiency. Given its increasing affordability and minimal subject burden, actigraphy should become a standard assessment tool in prospective studies of sleep and pain. In addition, ambulatory polysomnography is becoming more widely available and user-friendly, providing a unique opportunity to repeatedly assess electrophysiological sleep parameters within-person. Little is known about the longitudinal association of objective sleep disturbance and quantitative sensory tests of pain sensitivity or pain modulation. All of the prior studies relied on self-reported clinical pain. These questions should be taken up in future studies with both clinical and non-clinical samples.

Experimental Sleep Deprivation and Pain

Overview of Prior Work

Whereas recent advances in prospective studies have shed new light on the temporal relationship of sleep and pain, advances in sleep deprivation studies have provided deeper insights into the mechanisms by which sleep and pain are related. By manipulating specific aspects of a person's typical sleep opportunity period, these studies inform us of the impact of sleep loss on acute algesic responses to nociceptive stimuli, as well as any spontaneous changes in clinical pain. This topic has been the subject of a previous review 54, the results of which provided tentative support for the notion that sleep deprivation increases pain sensitivity. Out of 8 studies reviewed, 5 52,58,66,67,80 found evidence in favor of a hyperalgesic effect of sleep deprivation, and 3 2,22,79 failed to find evidence for sleep deprivation-induced hyperalgesia. The findings from those initial studies were limited in several respects. First, sample sizes were all quite low (Ns ranging from 9-20), and were restricted to healthy subjects, thereby limiting generality to clinical populations. Second, the sleep deprivation techniques were limited to total sleep deprivation and selective sleep stage deprivation. The former tells us what happens when the brain and body are completely deprived of sleep. The latter assesses the effects of disrupting specific sleep stages, such as slow wave sleep or rapid eye movement (REM). Notably absent from the early studies reviewed by Lautenbacher et al. 54 were partial sleep deprivation techniques, which may offer novel and more ecologically valid insights on how sleep and pain are related in both the general population and clinical samples. Partial sleep deprivation approaches involve curtailing sleep or disrupting sleep. Finally, the quantitative sensory testing (QST) modalities of early studies were limited to tests of thermal and mechanical threshold and tolerance, and did not assess the influence of sleep deprivation on descending pain modulatory processes such as pain inhibition or facilitation. Recent experimental studies have addressed some of these gaps, though many remain. Here, we review recent studies that have employed partial sleep deprivation techniques in the investigation of sleep disturbance-induced hyperalgesia. Additionally, we review several recent experimental sleep studies that have included clinical samples.

Recent Studies of Partial Sleep Deprivation and Pain Sensitivity

Most people with clinical sleep impairments achieve some measure of sleep throughout a typical sleep opportunity period. Therefore, partial sleep deprivation paradigms, which restrict sleep for part, but not all of the sleep opportunity period, may come closer than total sleep deprivation designs to approximating the effects of common sleep problems on pain sensitivity. Indeed, healthy subjects (N = 22) report spontaneous bodily pain after 2 nights of partial (4 hrs) sleep deprivation, and this effect increases as the number of nights of partial sleep deprivation increases 40. A small within-person study of healthy subjects (N = 7) found that 4-hour continuous sleep restriction for 2 consecutive nights significantly decreased finger withdrawal latency to a noxious suprathreshold thermal stimulus 96, suggesting that partial sleep deprivation may produce hyperalgesia. A recent partial sleep deprivation study of rheumatoid arthritis patients underscored the clinical relevance of those findings 46. In a relatively large sample of healthy subjects (N = 27) and rheumatoid arthritis patients (N = 27), continuous 4-hour sleep restriction for only one night resulted in elevations in self-reported pain, fatigue, depression, and anxiety in rheumatoid arthritis patients but not controls. Importantly, disease-specific measures of pain severity and painful joint counts were elevated in the rheumatoid arthritis patients following partial sleep deprivation 46.

A small within-person partial sleep deprivation experiment provided evidence suggesting differential effects of partial sleep deprivation on subjective pain reports versus cortical activation during QST 118. Subjects were instructed to restrict themselves to 4 hours or less of sleep in their home environment for one night, and sleep duration was verified with actigraphy. The next day, subjects arrived at the laboratory for quantitative sensory testing involving laser pulses of radiant heat (the same protocol was administered on a separate occasion following uninterrupted sleep). Noxious stimuli were rated as significantly more painful following partial sleep deprivation compared to uninterrupted sleep, while cortical EEG activity in the insular and cingulate regions was attenuated. These results might suggest that the hyperalgesic effect of partial sleep deprivation is mediated by impairments in the descending pain modulatory systems, rather than an amplification of the ascending sensory pathways, though further research is needed to confirm this possibility.

Partial sleep deprivation paradigms that disrupt sleep continuity, rather than maintain wake continuously for a specified period (e.g., 4 hours), may provide an even closer experimental analog to sleep disturbance in chronic pain. The predominant sleep complaint reported by patients with chronic pain is multiple nocturnal awakenings due to pain-related arousals throughout the night 117. Smith et al. 102 developed a forced awakenings (FA) sleep continuity disruption paradigm to enhance the predictive validity of hypotheses regarding sleep disturbance-induced hyperalgesia. FA is a partial sleep deprivation paradigm through which individuals are awakened pseudorandomly each hour during an 8-hour sleep opportunity period. Seven awakenings are for a 20-minute interval, and one awakening is for an entire 60-minute interval. In total, subjects are awake for 200 minutes and permitted to sleep for 280 minutes. Results from the initial FA study 102 demonstrated that disruption of sleep continuity resulted in next-day spontaneous pain reports in otherwise healthy women (N = 10) compared to a group receiving restricted sleep opportunity (N = 10) who slept continuously for an equivilent amount of time as the FA group, and healthy controls who slept continuously for 8 hours (N= 12). Additionally, conditioned pain modulation (CPM)—a measure of endogenous opioid-mediated pain inhibition 129—was significantly reduced following the sleep continuity disruption relative to both other groups 102.

The deleterious effect of sleep continuity disturbance on pain inhibitory function has since been replicated in several clinical studies. In a polysomnography study of TMD patients, poor sleep efficiency was significantly associated with diminished CPM efficacy, or impaired pain inhibition 28. Self-reported sleep efficiency was also inversely correlated with CPM efficacy in fibromyalgia patients 82. Further, CPM efficacy is reduced in rheumatoid arthritis patients relative to healthy controls, and appears to be mediated by self-reported sleep disturbance in that patient group 56.

Recent Studies of Experimental Sleep Deprivation in Clinical Samples

The majority of experimental sleep deprivation studies continue to rely on healthy samples in order to minimize error variance and establish basic causal effects. However, several recent studies have advanced the science by examining the hyperalgesic effects of sleep deprivation in clinical samples. As described above, Irwin et al. 46 demonstrated that partial sleep deprivation altered both psychosocial symptoms and disease-specific markers of rheumatoid arthritis. Comparable effects have been reported in patients with gastroesophageal reflux disease (GERD) who were exposed to partial sleep deprivation 99. Relative to healthy controls, GERD patients showed significantly greater increases in symptom intensity ratings following an acid perfusion test designed to evoke GERD-like symptoms. The authors interpreted changes in symptom intensity as clinically-relevant evidence of hyperalgesia following partial sleep deprivation.

Kundermann et al. 51 conducted the first study of sleep deprivation-induced hyperalgesia in a sample of patients with major depressive disorder. Patients demonstrated reductions in heat pain threshold following total sleep deprivation, whereas detection thresholds (e.g., touch) remained unchanged. These findings replicated effects from non-clinical studies 52, and suggest that total sleep deprivation selectively augments the activity of nociceptive pathways without altering the perception of non-nociceptive somatosensory stimuli. This study is limited, however, by the absence of a control group against which to compare the increases in hyperalgesia following sleep deprivation.

Finally, Busch et al. 14 recently reported that clinical pain complaints increased, but thermal and pressure pain thresholds were unchanged following total sleep deprivation in patients with chronic somatoform disorder. In contrast— and similar to Kundermann et al.'s study 51 with depressed patients—mood significantly improved following sleep deprivation. The findings suggest that in patients with somatoform disorder, mood and pain symptoms may be differentially affected by sleep disturbance, and that the hyperalgesic nociceptive responses observed in sleep deprivation studies involving healthy subjects may not translate to this patient population.

A recent experimental study suggests that the extension of sleep in individuals with mild chronic sleep loss attenuates baseline pain sensitivity levels 95. Individuals with an average sleep onset latency on the Multiple Sleep Latency Test of less than 8 minutes were randomized to 4 nights of an extended 10 hour bedtime or 4 nights of habitual bedtimes. Compared to habitual sleep, extended sleep increased tolerance to a radiant heat stimulus. Further, increased pain tolerance (i.e., reduced pain sensitivity) significantly correlated with changes in the Multiple Sleep Latency Test. As individuals with mild chronic sleep loss may be at risk for elevated pain sensitivity, these findings suggest that early intervention to increase sleep time may prevent chronic deficits in pain regulatory function.

Summary of Recent Experimental Sleep Deprivation Studies

A variety of sleep deprivation paradigms have been employed to test the direct effect of sleep disturbance on pain sensitivity. Across studies, it is evident that experimental disturbance of sleep, even after a single night, has the potential to increase both clinical pain and responses to quantitative sensory tests, although effects may vary between healthy and clinical populations. Hyperalgesic effects following experimental disruption of sleep continuity may be particularly relevant to patients with chronic pain, as they have been shown to functionally alter key endogenous pain modulatory pathways known to increase vulnerability to central sensitization and persistent pain 102. A next step, modeled elegantly by the Irwin et al. 46 study, will be to disrupt sleep continuity in a clinical population to determine its effect on disease-specific measures of pain and disability.

Discussion

Future Directions: Biobehavioral Mechanisms of the Association of Sleep and Pain

The focus of research on the association of sleep and pain is beginning to shift to mechanisms. Whereas early work was principally concerned with establishing whether sleep and pain were consistently associated, future work will undoubtedly become more concerned with how sleep and pain are associated. Here we highlight three broad areas that we believe hold great promise in explaining how sleep and pain are related.

Dopaminergic Signaling

Dopamine (DA) is the principal neurotransmitter of the forebrain reward system, a complex network of mesolimbic and nigrostriatal circuitry underlying the human behavioral drive to pursue pleasure. DA is also integral to the promotion and maintenance of arousal states 68, for reviews, see: 69 and is, therefore, intimately tied to the regulation of sleep and wake 25,83,87. DA receptors are abundant in the ascending reticular activating system, including aspects of the raphe nuclei in the brainstem, a critical sleep modulation region 3,61. Foo and Mason 37 postulated that serotonergic raphe cells signaling alertness may become dysregulated in the course of chronic pain, and contribute to prolonged periods of sleep loss and greater disruption of sleep continuity. Given the abundance of DA receptors in that region of the brain stem, and the well-known interaction of serotonergic and dopaminergic neurotransmission 49, it is possible that pain-induced alterations in DA signaling may influence the raphe nuclei modulation of sleep and wake.

This possibility is underscored by evidence indicating patients with chronic facial pain and fibromyalgia, respectively 55,63, have reduced DA metabolite concentrations in the cerebrospinal fluid 11,57,97, and a reduced phasic DA response to noxious stimuli 100,128. But basic mechanistic studies are needed to establish how sleep disturbance alters the function of DA, and how that alteration may consequently influence pain sensitivity. For example, Volkow et al. 124,125 conducted two total sleep deprivation experiments with PET imaging that found evidence for reduced [11C]raclopride binding to D2/D3 receptors in the striatum and thalamus of healthy human subjects. The authors initially interpreted these results to indicate that sleep deprivation enhanced endogenous DA tone in order to combat the primary drive to sleep, and postulated that the suprachiasmatic nucleus, which innervates the striatum and mesencephalon, may regulate this homeostatic process. However, a more recent sleep deprivation experiment from their group 123 found that the effects of methylphenidate on D2 receptor binding were no different following sleep deprivation compared to uninterrupted sleep. As methylphenidate is known to block the DAT receptor, this finding suggests that sleep deprivation may downregulate D2/D3 receptors, rather than enhance endogenous DA tone. Clearly, more research is needed to clarify the exact manner in which sleep deprivation alters DA, and whether those changes are correlated with concurrent changes in pain sensitivity. In addition, more clinical studies are needed to establish the behavioral consequences of sleep disturbance on reward processing, and how those changes influence coping with chronic pain.

Opioidergic Signaling

An abundance of transdisciplinary research suggests that opioid peptides play a critical mediating role in descending pain modulatory systems 5,7,48. Compromised pain inhibitory capacity has been demonstrated in many idiopathic clinical pain conditions with prominent sleep disturbance components 53,84,109, such as fibromyalgia. Preclinical studies indicate that sleep deprivation dysregulates endogenous opioid systems and attenuates the analgesic efficacy of μ-opioid receptor agonists 72,121. Several pathways could contribute to diminished opioid analgesia following sleep disruption. Opioid receptors are located in multiple nuclei that actively regulate both sleep and pain 37, including the preoptic suprachiasmatic nuclei, which controls sleep-wake cycles 19 and the periaqueductal gray, which plays a major role in descending pain inhibition98. Animal studies have found that sleep deprivation alters μ and δ opioid receptor function in mesolimbic circuits31, diminishes basal endogenous opioid levels 86 and down regulates central opioid receptors32. A recent study with humans supports these preclinical findings by demonstrating that diminished codeine analgesia is correlated with daytime sleepiness 110. Sleep, however, was not assessed. At the epidemiologic level, sleep disturbance cross-sectionally correlates with opioid use 133, but most of these studies focused on sleep apnea and cannot address directionality. Two diary studies of burn injury survivors provide preliminary evidence that a night of poor sleep predicts next day increases in opioid consumption 91,92. One way to probe the mechanisms underlying sleep-dependent impairment to opioid pathways is to determine whether sleep disturbance reduces the effectiveness of exogenously administered opioids. Efforts to that end are under way in our laboratory.

Negative and Positive Affect

Mediation analyses have revealed that elevations in negative affect (e.g., moods or emotions) may explain a significant portion of variance in the directional associations between sleep disturbance and pain in non-clinically depressed samples. However, studies have varied with respect to how the sleep/pain pathways were specified, requiring clarification in future research. One study found that negative mood mediates the relationship between sleep and pain in a heterogeneous sample of patients with back pain, fibromyalgia, and facial pain 75. A separate study of fibromyalgia patients treated sleep as a mediator of the pain-depression pathway, finding that sleep quality mediates the relationship between pain and symptoms of depression 64. Finally, another study of fibromyalgia patients found evidence in support of a model in which pain was a mediator of the pathway from sleep impairment to depressive symptoms 44. This set of findings suggests that sleep, pain, and negative mood share variance, but provide little clarity regarding the temporal dynamics of their associations. Prospective research could help resolve this dilemma by comparing several competing mediational models involving sleep, negative affect, and pain in multiple samples of patients with chronic pain and sleep disturbance.

Chung and Tso 16 reported that pharmaceutical treatment of depression did not attenuate the effects of self-reported and polysomnographic sleep disturbance on pain symptoms observed during an acute depressive episode, suggesting that sleep and pain vary beyond the influence of depression. This possibility is supported by several studies showing that the association of insomnia symptoms and chronic pain remains after controlling for depression symptoms 9,122,127. Going forward, studies should strive for consistency in the measurement of negative affect and depression symptoms, as findings may vary as a function of assessment instrument. In general, there has been relatively little attention directed toward distinctions in measurement of affective states within the context of studies on sleep and pain. Affect may be considered an umbrella term that comprises both moods, which are longer duration, lower intensity affective states, and emotions, which are shorter duration, higher intensity affective states 38. Both moods and emotions are discreet feeling states and are distinct from depression and other clinical disorders, which indicate frank mental health impairment. Thus, measures that target affective states, such as the Positive and Negative Affect Schedule 126, or discreet emotions 4, may be more appropriate for delineating mechanisms of the association of sleep and pain than depressive symptom inventories, which tend to be diffuse and nonspecific.

In a similar vein, the maladaptive cognitive coping style known as pain catastrophizing may also be an interesting target in mechanistic research. Pain catastrophizing is generally modestly correlated with negative affect but is distinct from clinical anxiety or depression 114. A recent study from our group demonstrated that sleep disturbance partially mediated the associations of pain catastrophizing and pain, and pain-related interference in a sample (N= 214) of TMD patients 13. In this study, the tendency to report ruminative catastrophizing thoughts when experiencing pain was associated with poorer self-reported sleep and increased self-reported pain. Entered together, sleep disturbance significantly attenuated the association between pain catastrophizing and both pain and pain-related interference with function. Thus, these findings raise the possibility that treatments targeting pain catastrophizing may have ‘cross-pollinating’ effects on both sleep and pain-related outcomes. The mechanism by which sleep disturbance mediates the association of pain catastrophizing and pain remains to be determined. One possibility supported by previous research is that high pain catastrophizers may have generally poorer sleep due to an inability to silence intrusive pain-related thoughts prior to bed time 104.

Although many studies investigating the association of sleep and pain have incorporated negative or depressed affect into their investigation, none have evaluated the potentially important role of positive affect. Positive affect is psychometrically distinct from negative affect 101,126, and has been shown to buffer, or attenuate, the relation of negative affect and chronic pain 130-132. Although the gold-standard cognitive-behavioral interventions for chronic pain and insomnia are primarily focused on reducing negative affect 26,120, a nascent but growing body of work has demonstrated that positive affect promotes resilient physical and psychosocial functioning among individuals with chronic pain 30,81,113,119 and insomnia 136. Positive affect has been identified as a unique contributor to the variance in pain reports in patients with fibromyalgia, for whom sleep disturbance is highly prevalent 131. A study of rheumatoid arthritis and fibromyalgia patients found that sleep duration uniquely moderated the association of pain and positive affect 43. Specifically, patients reporting 8 or more hours of sleep maintained positive affect through fluctuating pain states, whereas patients reporting 7 or fewer hours of sleep per night were more likely to have reduced positive affect when pain was elevated 43. The extent to which positive affect, independent of negative affect, contributes variance to the association of sleep and pain is unclear and should be investigated in future longitudinal and experimental studies.

Future Directions: Sociodemographic Moderators of the Association of Sleep and Pain

A wealth of data suggests that sleep and pain vary according to the most prominent sociodemographic factors. Out of a large literature on individual differences in sleep and pain, the most salient findings are that both sleep disturbance 35,36,78 and pain 85 tend to increase with age; African-Americans (AA) exhibit worse objective and subjective sleep impairments 23,42,107 and greater clinical and experimental pain sensitivity than Caucasians 27,90; and females exhibit worse objective and subjective sleep impairments 15,60,134 and greater clinical and experimental pain sensitivity than males 33,65,89. It will be important to determine whether the effect of sleep on pain, and vice versa, is moderated by key demographic variables, such as age, race, or sex. There is limited recent evidence that such interactions may reveal important sources of variance in the relation of sleep and pain.

Zhang et al. 135 investigated insomnia, pain, and somatic symptoms in a large sample of male and female adolescents (N = 259) and adults (N = 256). Moderation analyses revealed that females with insomnia reported significantly greater pain and somatic symptoms than females without insomnia, whereas no such effect for insomnia was observed among males. In fact, females without insomnia were comparable to males without insomnia, suggesting that insomnia may be a mechanism for the oft-noted sex differences in pain sensitivity 135. Further, although a similar pattern was observed as a statistical trend for adolescents, the effect was much stronger in adults, suggesting that sex differences associated with pain and insomnia may evolve with age. These findings highlight the rich opportunities for enhancing our understanding of the association of sleep and pain that emerge through sociodemographic moderation analyses. For example, given that AA report both greater sleep impairments and greater pain sensitivity, it is possible that the deleterious effects of sleep disturbance on pain are magnified in AA compared to Caucasians. Alternatively, it is possible that the increased clinical and experimental pain sensitivity in AA is better attributed to elevated sleep disturbance than to putative sociocultural influences on pain sensitivity. Such hypotheses should be tested by modelling interactions akin to those reported by Zhang et al. 135 in future studies.

General Summary and Conclusions

This review summarizes the recent longitudinal and experimental data on the association of sleep and pain. Due to the diversity of studies and methods, we chose to conduct a narrative review in order to compare and contrast across a wide range of studies. Still, due to the breadth of literature on sleep and pain, it was necessary to limit the scope of this review, which prevented us from discussing interesting studies on related topics, such as the role of sleep in inflammation 39,41. Our review is further limited by its inability to generate objective quantitative summary statistics, which are typically obtained within a more narrow and rigid scope of studies than we elected to review here. That being said, our review has revealed several important findings regarding the state of the science on sleep and pain.

First, over the past decade, the volume of studies investigating the association of sleep and pain has sharply risen. At the same time, methodological advances in study design, measurement, and analysis have unveiled a rich landscape of longitudinal and experimental effects. Large, longitudinal cohort studies with basic subjective assessments of sleep and pain generally support a reciprocal relationship between sleep disturbance (e.g., insomnia symptoms) and clinical pain reports. However, several longitudinal studies convincingly demonstrate that insomnia symptoms significantly increase the risk of developing future chronic pain disorders in previously pain-free individuals, whereas existing pain is not a strong predictor of new incident cases of insomnia 10,62. Further, deeper assessments of sleep disturbance and pain across multiple temporal sequences (e.g., microlongitudinal vs. longitudinal) tend to suggest that sleep disturbance is a stronger predictor of future pain than pain of sleep disturbance. These findings should encourage future investigators to incorporate a diverse set of pain and sleep assessments when feasible. Additionally, the longitudinal and microlongitudinal findings suggest that efforts to prevent and treat chronic pain may be well served to target sleep disturbance as a point of primary prevention and intervention.

It remains to be determined if the association of sleep and pain varies across different chronic pain disorders. The present review synthesizes findings from a wide range of disorders, including neuropathic, musculoskeletal, headache/migraine, and idiopathic pain disorders. The extent to which sleep disturbance differentially influences pain across disorder types may best be determined through mechanistic studies that identify reliable substrates of the sleep-pain dynamic. For example, current efforts in our lab and elsewhere to explicate the role of inflammation in the association of sleep disturbance and pain may yield data that is relevant for identifying which disorders are more likely to be differentially affected by sleep disturbance.

The experimental literature has expanded to include several studies of partial sleep deprivation and sleep continuity disruption that offer greater ecological validity than prior experimental designs. Combined with advances in quantitative sensory testing, these studies demonstrate that sleep disturbance may impair pain processing at multiple levels of the neuraxis, including those that regulate descending pain modulation.

Investigating the influence of biopsychosocial variables, such as positive and negative affect, brain dopamine and opioid systems, age, ethnicity, and sex on the relationship of sleep and pain may reveal mechanisms that can be targeted in novel treatments of patients with comorbid insomnia and chronic pain. The biopsychosocial model provides an excellent framework through which to integrate these and other potential mechanisms of the association of sleep and pain, such as inflammation 106 and neuroendocrine factors 108. Experimental studies that manipulate both sleep and pain are needed to isolate real-time changes in dopaminergic and opioidergic neurotransmission (e.g., through positron emission tomography and pharmacological challenge). Likewise, we are in need of a greater volume of microlongitudinal studies that take advantage of the increasingly feasible smartphone applications and ambulatory electroencephalogram technologies. Such studies are ideally suited for evaluation of biopsychosocial hypotheses of the association of sleep and pain, given their ability to assess dynamic changes in sleep architecture, transient pain flares, affect regulation, and cognitive coping, both within-person and across diverse social and ethnic community structures.

By testing mediation and moderation models of the biopsychosocial antecedents and sequelae of the association of sleep and pain, we may come closer the realizing the ultimate goals of research in this arena: improved clinical care of patients with sleep disorders and comorbid chronic pain. Integral to the optimization of clinical care is a better understanding of symptom trajectories that define not only who is most likely to develop poor outcomes, but when transitions to poor health are most likely to occur. Such a multilevel framework is critical to solving some of the most vexing clinical problems of our time, including the development of chronic pain following surgery, the experience of pain in relapsing/remitting conditions such as rheumatoid arthritis, lupus, and multiple sclerosis. Many of the tools required to pursue these questions are currently available, but we need to be asking the right questions in order to implement them effectively. The past decade of research has allowed us to move past the question of whether sleep and pain are related. Now, we must turn our attention to the biological, psychological, and social contingencies that qualify their association.

Acknowledgments

Disclosures: Funding for work on the current study was provided by NIH R01 AR05487 (MTS), NIH R01AR059410 (MTS) and NIH T32 NS070201 (PHF).

Footnotes

The authors declare no conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Affleck G, Urrows S, Tennen H, Higgins P, Abeles M. Sequential daily relations of sleep, pain intensity, and attention to pain among women with fibromyalgia. Pain. 1996;68:363–8. doi: 10.1016/s0304-3959(96)03226-5. [DOI] [PubMed] [Google Scholar]
  • 2.Arima T, Svensson P, Rasmussen C, Nielsen KD, Drewes AM, Arendt-Nielsen L. The relationship between selective sleep deprivation, nocturnal jaw-muscle activity and pain in healthy men. J Oral Rehabil. 2001;28:140–8. doi: 10.1046/j.1365-2842.2001.00687.x. [DOI] [PubMed] [Google Scholar]
  • 3.Bannon MJ, Roth RH. Pharmacology of mesocortical dopamine neurons. Pharm Rev. 1983;35:68. [PubMed] [Google Scholar]
  • 4.Barrett LF. Discrete emotions or dimensions? The role of valence focus and arousal focus. Cognition & Emotion. 1998;12:579–99. [Google Scholar]
  • 5.Basbaum AI, Fields HL. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu Rev Neurosci. 1984;7:309–38. doi: 10.1146/annurev.ne.07.030184.001521. [DOI] [PubMed] [Google Scholar]
  • 6.Beaulieu P, Walczak J. Pharmacological management of sleep and pain interactions. Sleep and Pain. 2007:391–416. [Google Scholar]
  • 7.Bencherif B, Fuchs PN, Sheth R, Dannals RF, Campbell JN, Frost JJ. Pain activation of human supraspinal opioid pathways as demonstrated by [11C]-carfentanil and positron emission tomography (PET) Pain. 2002;99:589–98. doi: 10.1016/S0304-3959(02)00266-X. [DOI] [PubMed] [Google Scholar]
  • 8.Bigatti SM, Hernandez AM, Cronan TA, Rand KL. Sleep disturbances in fibromyalgia syndrome: relationship to pain and depression. Arthritis Rheum. 2008;59:961–7. doi: 10.1002/art.23828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Boardman HF, Thomas E, Millson DS, Croft PR. Psychological, sleep, lifestyle, and comorbid associations with headache. Headache. 2005;45:657–69. doi: 10.1111/j.1526-4610.2005.05133.x. [DOI] [PubMed] [Google Scholar]
  • 10.Boardman HF, Thomas E, Millson DS, Croft PR. The natural history of headache: predictors of onset and recovery. Cephalalgia. 2006;26:1080–8. doi: 10.1111/j.1468-2982.2006.01166.x. [DOI] [PubMed] [Google Scholar]
  • 11.Bouckoms AJ, Sweet WH, Poletti C, Lavori P, Carr D, Matson W, Gamache P, Aronin N. Monoamines in the brain cerebrospinal fluid of facial pain patients. Anesth Prog. 1992;39:201–8. [PMC free article] [PubMed] [Google Scholar]
  • 12.Bromberg MH, Gil KM, Schanberg LE. Daily sleep quality and mood as predictors of pain in children with juvenile polyarticular arthritis. Health Psychol. 2012;31:202. doi: 10.1037/a0025075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Buenaver LF, Quartana PJ, Grace EG, Sarlani E, Simango M, Edwards RR, Haythornthwaite JA, Smith MT. Evidence for indirect effects of pain catastrophizing on clinical pain among myofascial temporomandibular disorder participants: The mediating role of sleep disturbance. Pain. 2012;153:1159–66. doi: 10.1016/j.pain.2012.01.023. [DOI] [PubMed] [Google Scholar]
  • 14.Busch V, Haas J, Cronlein T, Pieh C, Geisler P, Hajak G, Eichhammer P. Sleep deprivation in chronic somatoform pain-effects on mood and pain regulation. Psychiatry Res. 2012;195:134–43. doi: 10.1016/j.psychres.2011.07.021. [DOI] [PubMed] [Google Scholar]
  • 15.Buysse DJ, Germain A, Hall ML, Moul DE, Nofzinger EA, Begley A, Ehlers CL, Thompson W, Kupfer DJ. EEG spectral analysis in primary insomnia: NREM period effects and sex differences. Sleep. 2008;31:1673. doi: 10.1093/sleep/31.12.1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chung KF, Tso KC. Relationship between insomnia and pain in major depressive disorder: A sleep diary and actigraphy study. Sleep Med. 2010;11:752–8. doi: 10.1016/j.sleep.2009.09.005. [DOI] [PubMed] [Google Scholar]
  • 17.Davies KA, Macfarlane GJ, Nicholl BI, Dickens C, Morriss R, Ray D, McBeth J. Restorative sleep predicts the resolution of chronic widespread pain: results from the EPIFUND study. Rheumatology (Oxford) 2008;47:1809–13. doi: 10.1093/rheumatology/ken389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Davies M, Brophy S, Williams R, Taylor A. The prevalence, severity, and impact of painful diabetic peripheral neuropathy in type 2 diabetes. Diabetes Care. 2006;29:1518–22. doi: 10.2337/dc05-2228. [DOI] [PubMed] [Google Scholar]
  • 19.Desjardins GC, Brawer JR, Beaudet A. Distribution of mu, delta, and kappa opioid receptors in the hypothalamus of the rat. Brain Res. 1990;536:114–23. doi: 10.1016/0006-8993(90)90015-4. [DOI] [PubMed] [Google Scholar]
  • 20.Doufas AG, Panagiotou OA, Ioannidis JP. Concordance of Sleep and Pain Outcomes of Diverse Interventions: An Umbrella Review. PloS one. 2012;7:e40891. doi: 10.1371/journal.pone.0040891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Drewes AM, Nielsen KD, Hansen B, Taagholt SJ, Bjerregard K, Svendsen L. A longitudinal study of clinical symptoms and sleep parameters in rheumatoid arthritis. Rheumatology (Oxford) 2000;39:1287–9. doi: 10.1093/rheumatology/39.11.1287. [DOI] [PubMed] [Google Scholar]
  • 22.Drewes A, Rossel P, Arendt-Nielsen L, Nielsen KD, Hansen LM, Birket-Smith L, Stengaard-Pedersen K. Sleepiness does not modulate experimental joint pain in healthy volunteers. Scand J Rheumatol. 1997;26:399–400. doi: 10.3109/03009749709065709. [DOI] [PubMed] [Google Scholar]
  • 23.Durrence HH, Lichstein KL. The sleep of African Americans: a comparative review. Behav Sleep Med. 2006;4:29–44. doi: 10.1207/s15402010bsm0401_3. [DOI] [PubMed] [Google Scholar]
  • 24.Dzierzewski JM, Williams JM, Roditi D, Marsiske M, McCoy K, McNamara J, Dautovich N, Robinson ME, McCrae CS. Daily variations in objective nighttime sleep and subjective morning pain in older adults with insomnia: Evidence of covariation over time. J Am Geriatr Soc. 2010;58:925–30. doi: 10.1111/j.1532-5415.2010.02803.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dzirasa K, Ribeiro S, Costa R, Santos LM, Lin SC, Grosmark A, Sotnikova TD, Gainetdinov RR, Caron MG, Nicolelis MA. Dopaminergic control of sleep and wake states. J Neurosci. 2006;26:10577–89. doi: 10.1523/JNEUROSCI.1767-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Edinger JD, Wohlgemuth WK, Radtke RA, Marsh GR, Quillian RE. Cognitive behavioral therapy for treatment of chronic primary insomnia: a randomized controlled trial. JAMA. 2001;285:1856–64. doi: 10.1001/jama.285.14.1856. [DOI] [PubMed] [Google Scholar]
  • 27.Edwards CL, Fillingim RB, Keefe F. Race, ethnicity and pain. Pain. 2001;94:133–7. doi: 10.1016/S0304-3959(01)00408-0. [DOI] [PubMed] [Google Scholar]
  • 28.Edwards RR, Grace E, Peterson S, Klick B, Haythornthwaite JA, Smith MT. Sleep continuity and architecture: Associations with pain-inhibitory processes in patients with temporomandibular joint disorder. Eur J Pain. 2009;13:1043–7. doi: 10.1016/j.ejpain.2008.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Edwards RR, Almeida DM, Klick B, Haythornthwaite JA, Smith MT. Duration of sleep contributes to next-day pain report in the general population. Pain. 2008;137:202–7. doi: 10.1016/j.pain.2008.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Evers AW, Zautra A, Thieme K. Stress and resilience in rheumatic diseases: a review and glimpse into the future. Nat Rev Rheumatol. 2011;7:409–15. doi: 10.1038/nrrheum.2011.80. [DOI] [PubMed] [Google Scholar]
  • 31.Fadda P, Martellotta MC, De Montis MG, Gessa GL, Fratta W. Dopamine D1 and opioid receptor binding changes in the limbic system of sleep deprived rats. Neurochem Int. 1992;20(Suppl):153S–6S. doi: 10.1016/0197-0186(92)90229-k. [DOI] [PubMed] [Google Scholar]
  • 32.Fadda P, Tortorella A, Fratta W. Sleep deprivation decreases mu and delta opioid receptor binding in the rat limbic system. Neurosci Lett. 1991;129:315–7. doi: 10.1016/0304-3940(91)90489-g. [DOI] [PubMed] [Google Scholar]
  • 33.Fillingim RB, Edwards RR, Powell T. The relationship of sex and clinical pain to experimental pain responses. Pain. 1999;83:419–25. doi: 10.1016/S0304-3959(99)00128-1. [DOI] [PubMed] [Google Scholar]
  • 34.Finan PH, Smith MT. The comorbidity of insomnia, chronic pain, and depression: Dopamine as a putative mechanism. Sleep Med Rev. 2012 doi: 10.1016/j.smrv.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Foley D, Ancoli-Israel S, Britz P, Walsh J. Sleep disturbances and chronic disease in older adults: results of the 2003 National Sleep Foundation Sleep in America Survey. J Psychosom Res. 2004;56:497–502. doi: 10.1016/j.jpsychores.2004.02.010. [DOI] [PubMed] [Google Scholar]
  • 36.Foley DJ, Monjan AA, Brown SL, Simonsick EM, Wallace RB, Blazer DG. Sleep complaints among elderly persons: an epidemiologic study of three communities. Sleep. 1995;18:425–32. doi: 10.1093/sleep/18.6.425. [DOI] [PubMed] [Google Scholar]
  • 37.Foo H, Mason P. Brainstem modulation of pain during sleep and waking. Sleep Med Rev. 2003;7:145–54. doi: 10.1053/smrv.2002.0224. [DOI] [PubMed] [Google Scholar]
  • 38.Gross JJ, Thompson RA. Emotion regulation: Conceptual foundations. 3. 2007. p. 24. [Google Scholar]
  • 39.Haack M, Lee E, Cohen DA, Mullington JM. Activation of the prostaglandin system in response to sleep loss in healthy humans: potential mediator of increased spontaneous pain. Pain. 2009;145:136–41. doi: 10.1016/j.pain.2009.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Haack M, Mullington JM. Sustained sleep restriction reduces emotional and physical well-being. Pain. 2005;119:56–64. doi: 10.1016/j.pain.2005.09.011. [DOI] [PubMed] [Google Scholar]
  • 41.Haack M, Sanchez E, Mullington JM. Elevated inflammatory markers in response to prolonged sleep restriction are associated with increased pain experience in healthy volunteers. Sleep. 2007;30:1145. doi: 10.1093/sleep/30.9.1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hall MH, Matthews KA, Kravitz HM, Gold EB, Buysse DJ, Bromberger JT, Owens JF, Sowers M. Race and financial strain are independent correlates of sleep in midlife women: the SWAN sleep study. Sleep. 2009;32:73. [PMC free article] [PubMed] [Google Scholar]
  • 43.Hamilton NA, Catley D, Karlson C. Sleep and the affective response to stress and pain. Health Psychol. 2007;26:288–95. doi: 10.1037/0278-6133.26.3.288. [DOI] [PubMed] [Google Scholar]
  • 44.Hamilton NA, Pressman M, Lillis T, Atchley R, Karlson C, Stevens N. Evaluating Evidence for the Role of Sleep in Fibromyalgia: A Test of the Sleep and Pain Diathesis Model. Cognitive Therapy and Research. 2011:1–9. doi: 10.1007/s10608-011-9421-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Heo M, Allison DB, Faith MS, Zhu S, Fontaine KR. Obesity and quality of life: mediating effects of pain and comorbidities. Obes Res. 2012;11:209–16. doi: 10.1038/oby.2003.33. [DOI] [PubMed] [Google Scholar]
  • 46.Irwin MR, Olmstead R, Carrillo C, Sadeghi N, Fitzgerald JD, Ranganath VK, Nicassio PM. Sleep loss exacerbates fatigue, depression, and pain in rheumatoid arthritis. Sleep. 2012;35:537. doi: 10.5665/sleep.1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jansson Frojmark M, Boersma K. Bidirectionality between pain and insomnia symptoms: A prospective study. Br J Health Psychol. 2011;17:420–31. doi: 10.1111/j.2044-8287.2011.02045.x. [DOI] [PubMed] [Google Scholar]
  • 48.Julien N, Marchand S. Endogenous pain inhibitory systems activated by spatial summation are opioid-mediated. Neurosci Lett. 2006;401:256–60. doi: 10.1016/j.neulet.2006.03.032. [DOI] [PubMed] [Google Scholar]
  • 49.Kapur S, Remington G. Serotonin-dopamine interaction and its relevance to schizophrenia. Am J Psychiatry. 1996;153:466–76. doi: 10.1176/ajp.153.4.466. [DOI] [PubMed] [Google Scholar]
  • 50.Knutson KL, Ryden AM, Mander BA, Van Cauter E. Role of sleep duration and quality in the risk and severity of type 2 diabetes mellitus. Arch Intern Med. 2006;166:1768. doi: 10.1001/archinte.166.16.1768. [DOI] [PubMed] [Google Scholar]
  • 51.Kundermann B, Hemmeter-Spernal J, Huber MT, Krieg JC, Lautenbacher S. Effects of total sleep deprivation in major depression: overnight improvement of mood is accompanied by increased pain sensitivity and augmented pain complaints. Psychosom Med. 2008;70:92–101. doi: 10.1097/PSY.0b013e31815c1b5d. [DOI] [PubMed] [Google Scholar]
  • 52.Kundermann B, Spernal J, Huber MT, Krieg JC, Lautenbacher S. Sleep deprivation affects thermal pain thresholds but not somatosensory thresholds in healthy volunteers. Psychosom Med. 2004;66:932–7. doi: 10.1097/01.psy.0000145912.24553.c0. [DOI] [PubMed] [Google Scholar]
  • 53.Lautenbacher S, Rollman GB. Possible deficiencies of pain modulation in fibromyalgia. Clin J Pain. 1997;13:189–96. doi: 10.1097/00002508-199709000-00003. [DOI] [PubMed] [Google Scholar]
  • 54.Lautenbacher S, Kundermann B, Krieg JC. Sleep deprivation and pain perception. Sleep Med Rev. 2006;10:357–69. doi: 10.1016/j.smrv.2005.08.001. [DOI] [PubMed] [Google Scholar]
  • 55.Lee YC, Nassikas NJ, Clauw DJ. The role of the central nervous system in the generation and maintenance of chronic pain in rheumatoid arthritis, osteoarthritis and fibromyalgia. Arthritis Res Ther. 2011;13:211. doi: 10.1186/ar3306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lee YC, Lu B, Edwards RR, Wasan AD, Nassikas NJ, Clauw DJ, Solomon DH, Karlson EW. The role of sleep problems in central pain processing in rheumatoid arthritis. Arthritis Rheum. 2013;65:59–68. doi: 10.1002/art.37733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Legangneux E, Mora JJ, Spreux-Varoquaux O, Thorin I, Herrou M, Alvado G, Gomeni C. Cerebrospinal fluid biogenic amine metabolites, plasma-rich platelet serotonin and [3H]imipramine reuptake in the primary fibromyalgia syndrome. Rheumatology (Oxford) 2001;40:290–6. doi: 10.1093/rheumatology/40.3.290. [DOI] [PubMed] [Google Scholar]
  • 58.Lentz MJ, Landis CA, Rothermel J, Shaver JL. Effects of selective slow wave sleep disruption on musculoskeletal pain and fatigue in middle aged women. The Journal of rheumatology. 1999;26:1586. [PubMed] [Google Scholar]
  • 59.Lewandowski AS, Palermo TM, De la Motte S, Fu R. Temporal daily associations between pain and sleep in adolescents with chronic pain versus healthy adolescents. Pain. 2010;151:220. doi: 10.1016/j.pain.2010.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Li RHY, Wing YK, Ho SC, Fong SYY. Gender differences in insomnia--a study in the Hong Kong Chinese population. J Psychosom Res. 2002;53:601–9. doi: 10.1016/s0022-3999(02)00437-3. [DOI] [PubMed] [Google Scholar]
  • 61.Lu J, Jhou TC, Saper CB. Identification of wake-active dopaminergic neurons in the ventral periaqueductal gray matter. J Neurosci. 2006;26:193–202. doi: 10.1523/JNEUROSCI.2244-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lyngberg AC, Rasmussen BK, Jorgensen T, Jensen R. Has the prevalence of migraine and tension-type headache changed over a 12-year period? A Danish population survey. Eur J Epidemiol. 2005;20:243–9. doi: 10.1007/s10654-004-6519-2. [DOI] [PubMed] [Google Scholar]
  • 63.Meeus M, Nijs J. Central sensitization: a biopsychosocial explanation for chronic widespread pain in patients with fibromyalgia and chronic fatigue syndrome. Clin Rheumatol. 2007;26:465–73. doi: 10.1007/s10067-006-0433-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Miro E, Martinez MP, Sanchez AI, Prados G, Medina A. When is pain related to emotional distress and daily functioning in fibromyalgia syndrome? The mediating roles of self-efficacy and sleep quality. Br J Health Psychol. 2011;16:799–814. doi: 10.1111/j.2044-8287.2011.02016.x. [DOI] [PubMed] [Google Scholar]
  • 65.Mogil JS. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci. 2012;13:859–66. doi: 10.1038/nrn3360. [DOI] [PubMed] [Google Scholar]
  • 66.Moldofsky H, Scarisbrick P. Induction of neurasthenic musculoskeletal pain syndrome by selective sleep stage deprivation. Psychosom Med. 1976;38:35–44. doi: 10.1097/00006842-197601000-00006. [DOI] [PubMed] [Google Scholar]
  • 67.Moldofsky H, Scarisbrick P, England R, Smythe H. Musculosketal symptoms and non-REM sleep disturbance in patients with“ fibrositis syndrome” and healthy subjects. Psychosom Med. 1975;37:341–51. doi: 10.1097/00006842-197507000-00008. [DOI] [PubMed] [Google Scholar]
  • 68.Monti JM, Jantos h. The roles of dopamine and serotonin, and of their receptors, in regulating sleep and waking. Prog Brain Res. 2008;172:625–46. doi: 10.1016/S0079-6123(08)00929-1. [DOI] [PubMed] [Google Scholar]
  • 69.Monti JM, Monti D. The involvement of dopamine in the modulation of sleep and waking. Sleep Med Rev. 2007;11:113–33. doi: 10.1016/j.smrv.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 70.Morin CM, LeBlanc M, Daley M, Gregoire JP, Merette C. Epidemiology of insomnia: prevalence, self-help treatments, consultations, and determinants of help-seeking behaviors. Sleep Med. 2006;7:123–30. doi: 10.1016/j.sleep.2005.08.008. [DOI] [PubMed] [Google Scholar]
  • 71.Mork PJ, Nilsen TI. Sleep problems and risk of fibromyalgia: Longitudinal data on an adult female population in Norway. Arthritis Rheum. 2012;64:281–4. doi: 10.1002/art.33346. [DOI] [PubMed] [Google Scholar]
  • 72.Nascimento DC, Andersen ML, Hipolide DC, Nobrega JN, Tufik S. Pain hypersensitivity induced by paradoxical sleep deprivation is not due to altered binding to brain micro-opioid receptors. Behav Brain Res. 2007;178:216–20. doi: 10.1016/j.bbr.2006.12.016. [DOI] [PubMed] [Google Scholar]
  • 73.Nicassio PM, Wallston KA. Longitudinal relationships among pain, sleep problems, and depression in rheumatoid arthritis. J Abnorm Psychol. 1992;101:514. doi: 10.1037//0021-843x.101.3.514. [DOI] [PubMed] [Google Scholar]
  • 74.Nitter AK, Pripp AH, Forseth K. Are sleep problems and non-specific health complaints risk factors for chronic pain? A prospective population-based study with 17 year follow-up. Scandinavian Journal of Pain. 2012;3:210–7. doi: 10.1016/j.sjpain.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 75.O'Brien EM, Waxenberg LB, Atchison JW, Gremillion HA, Staud RM, McCrae CS, Robinson ME. Negative mood mediates the effect of poor sleep on pain among chronic pain patients. Clin J Pain. 2010;26:310–9. doi: 10.1097/AJP.0b013e3181c328e9. [DOI] [PubMed] [Google Scholar]
  • 76.O'Brien EM, Waxenberg LB, Atchison JW, Gremillion HA, Staud RM, McCrae CS, Robinson ME. Intraindividual variability in daily sleep and pain ratings among chronic pain patients: bidirectional association and the role of negative mood. Clin J Pain. 2011;27:425–33. doi: 10.1097/AJP.0b013e318208c8e4. [DOI] [PubMed] [Google Scholar]
  • 77.Odegard SS, Sand T, Engstrom M, Stovner LJ, Zwart JA, Hagen K. The Long-Term Effect of Insomnia on Primary Headaches: A Prospective Population-Based Cohort Study (HUNT-2 and HUNT-3) Headache: The Journal of Head and Face Pain. 2011;51:570–80. doi: 10.1111/j.1526-4610.2011.01859.x. [DOI] [PubMed] [Google Scholar]
  • 78.Ohayon MM, Carskadon MA, Guilleminault C, Vitiello MV. Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the human lifespan. Sleep. 2004;27:1255–73. doi: 10.1093/sleep/27.7.1255. [DOI] [PubMed] [Google Scholar]
  • 79.Older SA, Battafarano DF, Danning CL, Ward JA, Grady EP, Derman S, Russell IJ. The effects of delta wave sleep interruption on pain thresholds and fibromyalgia-like symptoms in healthy subjects; correlations with insulin-like growth factor I. The Journal of rheumatology. 1998;25:1180. [PubMed] [Google Scholar]
  • 80.Onen SH, Alloui A, Gross A, Eschallier A, Dubray C. The effects of total sleep deprivation, selective sleep interruption and sleep recovery on pain tolerance thresholds in healthy subjects. J Sleep Res. 2001;10:35–42. doi: 10.1046/j.1365-2869.2001.00240.x. [DOI] [PubMed] [Google Scholar]
  • 81.Ong AD, Zautra AJ, Reid MC. Psychological resilience predicts decreases in pain catastrophizing through positive emotions. Psychol Aging. 2010;25:516–23. doi: 10.1037/a0019384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Paul-Savoie E, Marchand S, Morin M, Bourgault P, Brissette N, Rattanavong V, Cloutier C, Bissonnette A, Potvin S. Is the deficit in pain inhibition in fibromyalgia influenced by sleep impairments? The open rheumatology journal. 2012;6:296. doi: 10.2174/1874312901206010296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Perogamvros L, Schwartz S. The roles of the reward system in sleep and dreaming. Neuroscience & Biobehavioral Reviews. 2012 doi: 10.1016/j.neubiorev.2012.05.010. [DOI] [PubMed] [Google Scholar]
  • 84.Peters ML, Schmidt AJ, Van den Hout MA, Koopmans R, Sluijter ME. Chronic back pain, acute postoperative pain and the activation of diffuse noxious inhibitory controls (DNIC) Pain. 1992;50:177–87. doi: 10.1016/0304-3959(92)90159-9. [DOI] [PubMed] [Google Scholar]
  • 85.Picavet HSJ, Hazes JMW. Prevalence of self reported musculoskeletal diseases is high. Ann Rheum Dis. 2003;62:644–50. doi: 10.1136/ard.62.7.644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Przewlocka B, Mogilnicka E, Lason W, van Luijtelaar EL, Coenen AM. Deprivation of REM sleep in the rat and the opioid peptides beta-endorphin and dynorphin. Neurosci Lett. 1986;70:138–42. doi: 10.1016/0304-3940(86)90452-0. [DOI] [PubMed] [Google Scholar]
  • 87.Qiu MH, Liu W, Qu WM, Urade Y, Lu J, Huang ZL. The Role of Nucleus Accumbens Core/Shell in Sleep-Wake Regulation and their Involvement in Modafinil-Induced Arousal. PloS one. 2012;7:e45471. doi: 10.1371/journal.pone.0045471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Quartana PJ, Wickwire EM, Klick B, Grace E, Smith MT. Naturalistic changes in insomnia symptoms and pain in temporomandibular joint disorder: A cross-lagged panel analysis. Pain. 2010;149:325–31. doi: 10.1016/j.pain.2010.02.029. [DOI] [PubMed] [Google Scholar]
  • 89.Racine M, Tousignant-Laflamme Y, Kloda LA, Dion D, Dupuis G, Choiniere M. A systematic literature review of 10years of research on sex/gender and pain perception: Part 2: Do biopsychosocial factors alter pain sensitivity differently in women and men? Pain. 2012;153:619–35. doi: 10.1016/j.pain.2011.11.026. [DOI] [PubMed] [Google Scholar]
  • 90.Rahim-Williams B, Riley JL, III, Williams AK, Fillingim RB. A quantitative review of ethnic group differences in experimental pain response: Do biology, psychology, and culture matter? Pain Med. 2012;13:522–40. doi: 10.1111/j.1526-4637.2012.01336.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Raymond I, Ancoli-Israel S, Choiniere M. Sleep disturbances, pain and analgesia in adults hospitalized for burn injuries. Sleep Med. 2004;5:551–9. doi: 10.1016/j.sleep.2004.07.007. [DOI] [PubMed] [Google Scholar]
  • 92.Raymond I, Nielsen TA, Lavigne G, Manzini C, Choiniere M. Quality of sleep and its daily relationship to pain intensity in hospitalized adult burn patients. Pain. 2001;92:381–8. doi: 10.1016/S0304-3959(01)00282-2. [DOI] [PubMed] [Google Scholar]
  • 93.Raymond I, Nielsen TA, Lavigne G, Manzini C, Choiniere M. Quality of sleep and its daily relationship to pain intensity in hospitalized adult burn patients. Pain. 2001;92:381–8. doi: 10.1016/S0304-3959(01)00282-2. [DOI] [PubMed] [Google Scholar]
  • 94.Riley JL, Benson MB, Gremillion HA, Myers CD, Robinson ME, Smith CL, Jr, Waxenberg LB. Sleep disturbance in orofacial pain patients: pain-related or emotional distress? Cranio: the journal of craniomandibular practice. 2001;19:106. doi: 10.1080/08869634.2001.11746159. [DOI] [PubMed] [Google Scholar]
  • 95.Roehrs TA, Harris E, Randall S, Roth T. Pain sensitivity and recovery from mild chronic sleep loss. Sleep. 2012;35:1667–72. doi: 10.5665/sleep.2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Roehrs T, Hyde M, Blaisdell B, Greenwald M, Roth T. Sleep loss and REM sleep loss are hyperalgesic. Sleep. 2006;29:145. doi: 10.1093/sleep/29.2.145. [DOI] [PubMed] [Google Scholar]
  • 97.Russell IJ, Vaeroy H, Javors M, Nyberg F. Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis Rheum. 1992;35:550–6. doi: 10.1002/art.1780350509. [DOI] [PubMed] [Google Scholar]
  • 98.Sastre JP, Buda C, Kitahama K, Jouvet M. Importance of the ventrolateral region of the periaqueductal gray and adjacent tegmentum in the control of paradoxical sleep as studied by muscimol microinjections in the cat 1. Neuroscience. 1996;74:415–26. doi: 10.1016/0306-4522(96)00190-x. [DOI] [PubMed] [Google Scholar]
  • 99.Schey R, Dickman R, Parthasarathy S, Quan SF, Wendel C, Merchant J, Powers J, Han B, van Handel D, Fass R. Sleep deprivation is hyperalgesic in patients with gastroesophageal reflux disease. Gastroenterology. 2007;133:1787–95. doi: 10.1053/j.gastro.2007.09.039. [DOI] [PubMed] [Google Scholar]
  • 100.Scott DJ, Heitzeg MM, Koeppe RA, Stohler CS, Zubieta JK. Variations in the human pain stress experience mediated by ventral and dorsal basal ganglia dopamine activity. J Neurosci. 2006;26:10789–95. doi: 10.1523/JNEUROSCI.2577-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Smith BW, Zautra AJ. Vulnerability and resilience in women with arthritis: test of a two-factor model. J Consult Clin Psychol. 2008;76:799–810. doi: 10.1037/0022-006X.76.5.799. [DOI] [PubMed] [Google Scholar]
  • 102.Smith MT, Edwards RR, McCann UD, Haythornthwaite JA. The effects of sleep deprivation on pain inhibition and spontaneous pain in women. Sleep. 2007;30:494–505. doi: 10.1093/sleep/30.4.494. [DOI] [PubMed] [Google Scholar]
  • 103.Smith MT, Haythornthwaite JA. How do sleep disturbance and chronic pain inter-relate? Insights from the longitudinal and cognitive-behavioral clinical trials literature. Sleep Med Rev. 2004;8:119–32. doi: 10.1016/S1087-0792(03)00044-3. [DOI] [PubMed] [Google Scholar]
  • 104.Smith MT, Perlis ML, Carmody TP, Smith MS, Giles DE. Presleep cognitions in patients with insomnia secondary to chronic pain. J Behav Med. 2001;24:93–114. doi: 10.1023/a:1005690505632. [DOI] [PubMed] [Google Scholar]
  • 105.Smith MT, Klick B, Kozachik S, Edwards RE, Holavanahalli R, Wiechman S, Blakeney P, Lezotte D, Fauerbach JA. Sleep onset insomnia symptoms during hospitalization for major burn injury predict chronic pain. Pain. 2008;138:497. doi: 10.1016/j.pain.2008.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Smith MT, Quartana PJ, Okonkwo RM, Nasir A. Mechanisms by which sleep disturbance contributes to osteoarthritis pain: a conceptual model. Current pain and headache reports. 2009;13:447–54. doi: 10.1007/s11916-009-0073-2. [DOI] [PubMed] [Google Scholar]
  • 107.Song Y, Ancoli-Israel S, Lewis CE, Redline S, Harrison SL, Stone KL. The association of race/ethnicity with objectively measured sleep characteristics in older men. Behav Sleep Med. 2011;10:54–69. doi: 10.1080/15402002.2012.636276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Spath-Schwalbe E, Uthgenannt D, Voget G, Kern W, Born J, Fehm HL. Corticotropin-releasing hormone-induced adrenocorticotropin and cortisol secretion depends on sleep and wakefulness. Journal of Clinical Endocrinology & Metabolism. 1993;77:1170–3. doi: 10.1210/jcem.77.5.8077308. [DOI] [PubMed] [Google Scholar]
  • 109.Staud R, Robinson ME, Vierck CJ, Jr, Price DD. Diffuse noxious inhibitory controls (DNIC) attenuate temporal summation of second pain in normal males but not in normal females or fibromyalgia patients. Pain. 2003;101:167–74. doi: 10.1016/s0304-3959(02)00325-1. [DOI] [PubMed] [Google Scholar]
  • 110.Steinmiller CL, Roehrs TA, Harris E, Hyde M, Greenwald MK, Roth T. Differential effect of codeine on thermal nociceptive sensitivity in sleepy versus nonsleepy healthy subjects. Exp Clin Psychopharmacol. 2010;18:277–83. doi: 10.1037/a0018899. [DOI] [PubMed] [Google Scholar]
  • 111.Stepanski EJ, Walker MS, Schwartzberg LS, Blakely LJ, Ong JC, Houts AC. The relation of trouble sleeping, depressed mood, pain, and fatigue in patients with cancer. Journal of clinical sleep medicine: JCSM: official publication of the American Academy of Sleep Medicine. 2009;5:132. [PMC free article] [PubMed] [Google Scholar]
  • 112.Stone AA, Broderick JE, Porter LS, Kaell AT. The experience of rheumatoid arthritis pain and fatigue: examining momentary reports and correlates over one week. Arthritis Rheum. 1997;10:185–93. doi: 10.1002/art.1790100306. [DOI] [PubMed] [Google Scholar]
  • 113.Sturgeon JA, Zautra AJ. Resilience: a new paradigm for adaptation to chronic pain. Curr Pain Headache Rep. 2010;14:105–12. doi: 10.1007/s11916-010-0095-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sullivan MJ, Bishop SR, Pivik J. The pain catastrophizing scale: development and validation. Psychological assessment. 1995;7:524. [Google Scholar]
  • 115.Tang NK, Goodchild CE, Sanborn AN, Howard J, Salkovskis PM. Deciphering the temporal link between pain and sleep in a heterogeneous chronic pain patient sample: a multilevel daily process study. Sleep. 2012;35:675. doi: 10.5665/sleep.1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tang NK. Cognitive-behavioral therapy for sleep abnormalities of chronic pain patients. Curr Rheum Report. 2009;11:451–60. doi: 10.1007/s11926-009-0066-5. [DOI] [PubMed] [Google Scholar]
  • 117.Taylor DJ, Mallory LJ, Lichstein KL, Durrence HH, Riedel BW, Bush AJ. Comorbidity of chronic insomnia with medical problems. Sleep. 2007;30:213–8. doi: 10.1093/sleep/30.2.213. [DOI] [PubMed] [Google Scholar]
  • 118.Tiede W, Magerl W, Baumg+ñrtner U, Durrer B, Ehlert U, Treede RD. Sleep restriction attenuates amplitudes and attentional modulation of pain-related evoked potentials, but augments pain ratings in healthy volunteers. Pain. 2010;148:36–42. doi: 10.1016/j.pain.2009.08.029. [DOI] [PubMed] [Google Scholar]
  • 119.Tugade MM, Fredrickson BL, Barrett LF. Psychological resilience and positive emotional granularity: examining the benefits of positive emotions on coping and health. J Pers. 2004;72:1161–90. doi: 10.1111/j.1467-6494.2004.00294.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Turner JA, Holtzman S, Mancl L. Mediators, moderators, and predictors of therapeutic change in cognitive-behavioral therapy for chronic pain. Pain. 2007;127:276–86. doi: 10.1016/j.pain.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 121.Ukponmwan OE, Rupreht J, Dzoljic MR. REM sleep deprivation decreases the antinociceptive property of enkephalinase-inhibition, morphine and cold-water-swim. Gen Pharmacol. 1984;15:255–8. doi: 10.1016/0306-3623(84)90170-8. [DOI] [PubMed] [Google Scholar]
  • 122.Vgontzas A, Cui L, Merikangas KR. Are sleep difficulties associated with migraine attributable to anxiety and depression? Headache. 2008;48:1451–9. doi: 10.1111/j.1526-4610.2008.01175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Volkow ND, Tomasi D, Wang GJ, Telang F, Fowler JS, Logan J, Benveniste H, Kim R, Thanos PK, Ferre S. Evidence that sleep deprivation downregulates dopamine D2R in ventral striatum in the human brain. J Neurosci. 2012;32:6711–7. doi: 10.1523/JNEUROSCI.0045-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Volkow ND, Tomasi D, Wang GJ, Telang F, Fowler JS, Wang RN, Logan J, Wong C, Jayne M, Swanson JM. Hyperstimulation of striatal D2 receptors with sleep deprivation: Implications for cognitive impairment. Neuroimage. 2009;45:1232–40. doi: 10.1016/j.neuroimage.2009.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Volkow ND, Wang GJ, Telang F, Fowler JS, Logan J, Wong C, Ma J, Pradhan K, Tomasi D, Thanos PK, Ferre S, Jayne M. Sleep deprivation decreases binding of [11C]raclopride to dopamine D2/D3 receptors in the human brain. J Neurosci. 2008;28:8454–61. doi: 10.1523/JNEUROSCI.1443-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Watson D, Clark LA, Tellegen A. Development and validation of brief measures of positive and negative affect: the PANAS scales. J Pers Soc Psychol. 1988;54:1063–70. doi: 10.1037//0022-3514.54.6.1063. [DOI] [PubMed] [Google Scholar]
  • 127.Wilson KG, Eriksson MY, D'Eon JL, Mikail SF, Emery PC. Major depression and insomnia in chronic pain. Clin J Pain. 2002;18:77–83. doi: 10.1097/00002508-200203000-00002. [DOI] [PubMed] [Google Scholar]
  • 128.Wood PB, Schweinhardt P, Jaeger E, Dagher A, Hakyemez H, Rabiner EA, Bushnell MC, Chizh BA. Fibromyalgia patients show an abnormal dopamine response to pain. Eur J Neurosci. 2007;25:3576–82. doi: 10.1111/j.1460-9568.2007.05623.x. [DOI] [PubMed] [Google Scholar]
  • 129.Yarnitsky D. Conditioned pain modulation (the diffuse noxious inhibitory control-like effect): its relevance for acute and chronic pain states. Curr Opin Anaesthesiol. 2010;23:611–5. doi: 10.1097/ACO.0b013e32833c348b. [DOI] [PubMed] [Google Scholar]
  • 130.Zautra AJ, Affleck GG, Tennen H, Reich JW, Davis MC. Dynamic approaches to emotions and stress in everyday life: Bolger and Zuckerman reloaded with positive as well as negative affects. J Pers. 2005;73:1511–38. doi: 10.1111/j.0022-3506.2005.00357.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Zautra AJ, Fasman R, Reich JW, Harakas P, Johnson LM, Olmsted ME, Davis MC. Fibromyalgia: evidence for deficits in positive affect regulation. Psychosom Med. 2005;67:147–55. doi: 10.1097/01.psy.0000146328.52009.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zautra AJ, Johnson LM, Davis MC. Positive affect as a source of resilience for women in chronic pain. J Consult Clin Psychol. 2005;73:212–20. doi: 10.1037/0022-006X.73.2.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Zgierska A, Brown RT, Zuelsdorff M, Brown D, Zhang Z, Fleming MF. Sleep and daytime sleepiness problems among patients with chronic noncancerous pain receiving long-term opioid therapy: a cross-sectional study. J Opioid Manag. 2007;3:317–27. doi: 10.5055/jom.2007.0020. [DOI] [PubMed] [Google Scholar]
  • 134.Zhang B, Wing Y. Sex differences in insomnia: a meta-analysis. Sleep. 2006;29:85. doi: 10.1093/sleep/29.1.85. [DOI] [PubMed] [Google Scholar]
  • 135.Zhang J, Lam SP, Li SX, Tang NL, Yu MWM, Li AM, Wing YK. Insomnia, sleep quality, pain, and somatic symptoms: Sex differences and shared genetic components. Pain. 2012;153:666–73. doi: 10.1016/j.pain.2011.12.003. [DOI] [PubMed] [Google Scholar]
  • 136.Zohar D, Tzischinsky O, Epstein R, Lavie P. The effects of sleep loss on medical residents' emotional reactions to work events: a cognitive-energy model. Sleep. 2005;28:47–54. doi: 10.1093/sleep/28.1.47. [DOI] [PubMed] [Google Scholar]

RESOURCES