Your activity: 62 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Pathogenesis of fibromyalgia

Pathogenesis of fibromyalgia
Author:
Don L Goldenberg, MD
Section Editor:
Peter H Schur, MD
Deputy Editor:
Philip Seo, MD, MHS
Literature review current through: Dec 2022. | This topic last updated: Sep 29, 2021.

INTRODUCTION — The characteristic feature of fibromyalgia (FM) is chronic widespread pain in the absence of peripheral musculoskeletal inflammation or structural damage. FM may complicate many rheumatic diseases or occur independently. FM is only one of many causes of widespread pain. (See "Overview of chronic widespread (centralized) pain in the rheumatic diseases".)

There is no evidence that a single event "causes" FM. Rather, many physical and/or emotional stressors may trigger or aggravate symptoms. These have included certain infections, such as a viral illness or Lyme disease, as well as emotional or physical trauma [1,2].

The pathogenesis of fibromyalgia is presented here. A detailed description of the clinical manifestations of FM and an approach to the diagnosis and differential diagnosis of FM in adults and children are presented separately. (See "Clinical manifestations and diagnosis of fibromyalgia in adults" and "Fibromyalgia in children and adolescents: Clinical manifestations and diagnosis" and "Differential diagnosis of fibromyalgia".)

TERMINOLOGY AND PAIN CLASSIFICATION — Fibromyalgia (FM) is considered to be a disorder of pain regulation, often classified under the term central sensitization [1-3] (see 'Central nervous system altered pain processing' below). Central sensitization has been defined as an amplification of neural signaling within the central nervous system (CNS) resulting in pain hypersensitivity [3]. The International Association for the Study of Pain has suggested a new mechanism description, nociplastic pain, to encompass conditions characterized by altered pain sensitization [3].

During much of the 20th century, FM was thought to be a muscle disease or related to soft-tissue inflammation, hence the term fibrositis. However, controlled comparisons found no evidence for significant pathologic or biochemical muscle abnormalities [1,2,4-6]. As an example, measures of muscle function, including force generation and lactate production during exercise, and muscle pain following exertion are remarkably similar in women with FM and sedentary controls [5,6]. Soft-tissue tender points, initially part of the FM diagnostic criteria, represent CNS pain dysregulation rather than localized pathology [1-3].

FM shares several clinical and pathophysiologic features with other common pain disorders that are considered to be more central rather than peripheral pain conditions, such as migraine, tension headaches, temporomandibular joint disorder, and irritable bowel syndrome. Clinical characteristics of each of these conditions include widespread pain, fatigue, and sleep and mood disturbances. These conditions also share common genetic and CNS pain processing mechanisms with FM. More limited studies have suggested there might also be a role for peripheral neuropathic mechanisms or focal tissue changes in some patients. (See 'Peripheral pain mechanisms' below.)

GENETIC PREDISPOSITION AND CANDIDATE GENES — A number of observational and biologic studies suggest that chronic widespread pain and fibromyalgia (FM) have, in part, a genetic basis [7]. First-degree relatives of patients with FM are 8.5 times more likely to have FM than relatives of patients with rheumatoid arthritis [8]. Familial aggregation of lowered thresholds for pressure-induced pain has been documented in first-degree relatives of patients. Such reports suggest a shared hereditary factor that may account for the overlap of chronic pain and mood disorders in families. However, no clear association between chronic widespread pain and any single candidate gene has yet been conclusively documented [9].

The ability of some antidepressant drugs to improve symptoms suggested that genes involved in serotonin and/or catecholamine metabolic or signaling pathways might be candidates for conferring susceptibility. Pain-related genes that have been potentially associated with FM include those for catechol-O-methyltransferase (COMT), mu-opioid receptors, voltage-gated sodium channels, GTP cyclohydrolase 1, and gamma-aminobutyric acid (GABA)ergic pathways [7,9].

The first large candidate gene study evaluated 496 patients with FM and 348 chronic pain-free controls [10]. They evaluated >350 genes known to be involved in nociception, inflammation, and affect. Significant differences in allele frequencies between FM cases and controls were observed for three genes: GABRB3 (rs4906902, p = 3.65x10-6), TAAR1 (rs8192619, p = 1.11x10-5), and GBP1 (rs7911, p = 1.06x10-4). These three genes and seven other genes with suggestive evidence for association were examined in a second, independent cohort of patients with FM and controls genotyped using the Perlegen 600K platform. Evidence of association in the replication cohort was observed for TAAR1, RGS4, CNR1, and GRIA4 genes.

The first genome-wide linkage scan for FM was performed in a cohort of 116 families from the Fibromyalgia Family Study [11]. The estimated sibling recurrence risk ratio for FM was 13.6, based upon a reported FM population prevalence of 2 percent. Genome-wide suggestive evidence of linkage was observed at markers D17S2196 and D17S1294 on chromosome 17p11.2-q11.2. These markers have potential impact on various pain pathways.

A subsequent genome-wide profiling found that, compared with controls, patients with FM had differences in expression of 421 genes, many of which were important in pain processing [12]. They used diagnostic models and identified a subset of 10 gene signature sets that provided 95 percent sensitivity and specificity for FM.

A genome-wide association study of nearly 7000 patients with chronic widespread pain found an association with the RNF123 locus and possible association with the ATP2C1 locus, both involved in calcium regulation [13]. This study could not confirm the association with COMT in chronic widespread pain.

Experimental pain studies have demonstrated that the COMT polymorphism affects central pain [7]. COMT regulates the catabolism of catechol neurotransmitters, including epinephrine, norepinephrine, and dopamine. The frequency of genetic variations associated with low COMT enzyme activity was significantly higher in patients with FM than in healthy volunteers [14]. Patients with FM were more sensitive to experimental pain than healthy volunteers, and in particular, FM individuals with the Met/Met genotype (Val158Met SNP) or the HPS-APS haplotypes showed higher sensitivity to thermal and pressure pain stimuli than patients carrying the LPS haplotype or val alleles (Val158Met SNP). Another study demonstrated that the Met/Met genotype was significantly higher in patients with FM than healthy controls [15].A case-control study of more than 400 Korean patients with FM and 400 controls found that polymorphisms of the COMT gene correlated with the risk for FM as well as pain sensitivity [16]. However, other studies have not found a direct association of COMT haplotypes with FM, including one report of over 2700 patients with FM [17]. In that report, the minor COMT alleles were overrepresented, suggesting that COMT may indirectly influence FM.

Specific genes involved in the serotonin pathway also increase the risk of conditions like FM, although it has been difficult to determine whether this is a marker of psychological status or pain propensity [18]. Polymorphisms in the serotonin receptor gene (HTR1A) did not differ in patients with FM compared with controls and did not correlate with pain or other symptoms in patients with FM [19].

One report evaluated the A118G rs1799971 polymorphism in the opioid receptor mu 1 gene (OPRM1) in FM [20]. The 118G allele frequency was significantly lower in patients with FM than in the control group. The translocator protein gene (TPSO), which is upregulated during glial activation in chronic pain states, was associated with greater pain intensity as well as more FM symptoms [21]. These investigators then studied the effects of three functional genetic polymorphisms on exercise-induced hypoalgesia in 130 patients with FM and matched controls [22]. They found opposing interactions between opioid and serotonergic pathways. Polymorphisms of the u-opioid receptor gene were found to influence brain pain processing in patients with FM [23]. G-allele carriers demonstrated increased activation in the precentral gyrus but decreased functional connectivity with the frontal control network with painful stimuli.

Genotype frequencies in 314 Spanish women with FM were compared with controls [24]. Single nucleotide polymorphism associations of guanosine triphosphate cyclohydrolase 1, COMT, and opioid receptor (OPRM1) genes with FM were found. There has also been an association of FM with brain-derived neurotrophic factor (BDNF) gene polymorphisms [25].

A few studies have focused on genes other than those involved in pain pathways. One study using whole exome sequencing found evidence in their cohort of a gene variant in 13 percent of patients with FM that was associated with increased plasma levels of monocyte chemoattractant protein (MCP)-1 and interferon gamma-induced protein (IP)-10, compared with patients with FM who had the wild-type allele; and another variant in 11 percent of patients that was associated with elevated levels of interleukin (IL)-12 compared with patients carrying the wild-type [26].

Epigenetics, which involves the study of hereditary changes not attributable to alterations in DNA sequence, may provide new approaches to gene-environment interactions in conditions like FM. For example, DNA methylation and histone modification alter gene expression without changes in DNA. DNA methylation in patients with FM differed from healthy controls, including in genes involved in DNA repair and membrane transport [27]. The epigenetics of BDNF was studied in patients with FM and comorbid myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) [28]. The serum BDNF were elevated, and lower BDNF methylation predicted higher BDNF levels and correlated with patients' symptoms and widespread pain in the FM/ME/CFS subjects.

In a study of FM in 26,749 individuals undergoing elective surgery, younger patients with an FM phenotype had a stronger genetic component than older individuals [29]. Overall, the FM phenotype had an estimated heritability of 14 percent.

CENTRAL NERVOUS SYSTEM ALTERED PAIN PROCESSING — Multiple lines of evidence have demonstrated that fibromyalgia (FM) is a disorder of pain processing, including the following:

Temporal summation of pain – Patients with FM experience greater than normal increases in the perceived intensity of pain when rapidly repetitive short noxious stimuli are administered, which is termed temporal summation of pain [4,30].

Decreased endogenous pain inhibition – Endogenous analgesic systems appear to be deficient in FM [31]. There are both a reduction in diffuse noxious inhibitory control (in which decreased pain occurs upon stimulation with a second acutely painful stimulus) and an inability to inhibit irrelevant sensory stimuli following repetitive nonpainful stimulation [32,33].

Pain receptors and pain-related neuropeptides – Changes are seen in opioid receptors, including upregulation in the periphery and a reduction in the brain [34,35]. Substance P, a neuropeptide associated with chronic pain states, is increased in the cerebrospinal fluid compared with controls [36]. Increased brain and plasma brain-derived neurotrophic factor have been found in FM [37].

The increased pain sensitivity in FM is part of a generalized hypersensitivity to sensory stimuli, including to visual, auditory, and olfactory stimuli. Patients with FM, compared with healthy controls, have exhibited increased brain responses to both pain onset and pain offset [38]. Quantitative sensory testing (QST) demonstrated hypersensitivity to sound, as well as to heat and mechanical pain, in patients with FM compared with healthy controls [39].

BRAIN NEUROIMAGING

Heightened pain response to experimental pain stimulus — Functional magnetic resonance imaging (fMRI) demonstrated that patients with FM had greater neuronal activity in pain-processing brain regions compared with controls, following the same pressure stimuli [40]. Differences in activation of pain-sensitive areas of the brain have also been noted with fMRI [41]. Areas of the brain that consistently exhibit greater activation after the same stimulus in patients with FM than controls include the secondary somatosensory cortex, insula, and anterior cingulate cortex [41].

fMRI has also been utilized to demonstrate the important role of comorbid psychological factors on pain perception in patients with FM [42]; patients with FM and comorbid depression demonstrate increased cerebral blood flow in the amygdala and anterior insula, areas important in the affective pain response. However, unlike FM, depression does not seem to affect the level of neuronal activation in sensory pain regions such as the secondary somatosensory cortex.

Patients with FM exhibited less robust activations during both anticipation of pain and anticipation of relief within regions commonly thought to be involved in sensory, affective, cognitive, and pain-modulatory processes [43]. Reduced reward/punishment signaling in FM may be related to the augmented central processing of pain and reduced efficacy of opioid treatments in these patients.

Patients with FM showed a lack of pain reduction impact from a positive emotional context [44]. Compared with controls, there was less activation in the secondary somatosensory cortex, insula, orbitofrontal cortex, and anterior cingulate cortex during positive picture pain trials.

fMRI demonstrated that, compared with controls, patients with FM had greater neuronal activity during catastrophizing, particularly in regions of the posterior cingulate cortex [45]. However, an arterial spin labelling study found no significant differences in resting state blood flow in pain processing areas between patients with FM and healthy controls [46].

Changes in brain morphology — Morphometric analysis by MRI in patients with FM shows, compared with healthy controls, a significant reduction in total gray matter volume and a threefold increase in age-associated loss of gray matter, suggesting premature aging of the brain [47]. The degree of loss was greater in patients with a longer duration of disease. Such gray matter loss, which is also reported in other chronic pain and stress-related disorders, was most prominent in regions related to stress and pain processing but was also seen in areas related to cognitive function. Another study revealed similar findings [48]. However, a different group found no significant difference in gray matter between patients with FM and controls when controlling for depression [49]. Decreased gray matter in patients with FM was associated with T1 relaxation times, a marker of water content [50]. Regional brain gray matter increases were associated with GABAA receptor concentration, indicative of neuronal plasticity.

Structural changes and functional connectivity of the brain during application of intermittent pressure-pain stimuli were compared between 26 patients with FM and 13 age- and sex-matched healthy controls [51]. The rostral anterior cingulate cortex in the patients with FM exhibited decreased cortical thickness, brain volume, and regional functional connectivity compared with the controls. The structural changes correlated with duration of symptoms.

Diffusion weighted imaging has demonstrated white matter changes in FM that may be associated with alterations in pain intensity [52]. The FM group demonstrated lower fractional anisotropy in the left body of the corpus callosum. These values were negatively associated with sensory pain, suggesting disruption of white matter micro-structure and an association with clinical pain intensity.

A systematic review of imaging studies in FM found moderate evidence that central sensitization is correlated with a gray matter volume decrease in specific brain regions (mainly anterior cingulate cortex and prefrontal cortex) [53]. They found evidence of decreased functional connectivity in the descending pain-modulating system in patients with FM.

Altered neurotransmitter function — Using proton magnetic resonance (MR) spectroscopy, patients with FM had significantly higher levels of glutamine within the right posterior insula compared with controls [54]. Elevated insular glutamate in FM is associated with experimental pain. Within the right posterior insula, higher levels of glutamate were associated with lower pressure pain thresholds. More limited data, using positron emission tomography, have shown reduced dopaminergic activity in the response to pain in patients with FM compared with controls [55].

Patients with FM were evaluated for cortical excitability and intracortical modulation using transcranial magnetic stimulation of the motor cortex [56]. The patients with FM compared with controls had deficits in intracortical modulation of GABAergic and glutamatergic mechanisms. Using MR spectroscopy, patients with FM showed higher levels of glutamate and a higher glutamine-glutamate/creatine ratio in the right amygdala compared with controls [57]. In the patients with FM with more pain, fatigue and depressive symptoms inositol (Ins) levels were found to be significantly higher in the right amygdala and right thalamus.

Using proton MR spectroscopy, GABA levels in the right anterior insula were significantly lower in patients with FM compared with healthy controls [58]. No significant differences between groups were detected in the posterior insula or occipital cortex. Within the right posterior insula, higher levels of GABA were positively correlated with pressure-pain thresholds in the patients with FM.

Changes in resting-state functional connectivity — fMRI has been particularly useful in evaluating brain connectivity [41,59]. In one study, abnormal resting state functional connectivity of the periaqueductal gray was noted in FM subjects compared with controls [60]. The authors suggested that the changes result in impaired descending pain inhibition. Altered functional connectivity with the default mode network, a region active when the brain is at rest, and the insula, a key pain-processing region, was noted in FM subjects compared with controls [61]. Altered spinal cord neuronal activity between the ventral and dorsal spinal cord was found in FM but not in controls [62]. Resting-state fMRI demonstrated altered hub structure, regions that effectively transmit neural information, in FM compared with controls [63]. Altered hub topology within the insula was associated with clinical pain intensity. The neural organization of intrinsic functional brain hubs or communities in patients with FM differed from that of controls without FM, demonstrating decreased neural stability [64]. Changes in this neural organization correlated with levels of pain.

fMRI was utilized to identify a brain signature that may characterize FM by following responses to painful pressure and nonpainful multisensory stimuli [2]. Specific pain-related multisensory patterns classified patients with FM versus controls with 92 percent sensitivity and 94 percent specificity. The neuroimaging brain response to pain-related fear was explored as a brain signature of FM and found to be a predictor of FM as well as its treatment response [65].

SLEEP/MOOD/COGNITIVE ABNORMALITIES — Underlying central nervous system (CNS) dysfunction is suggested by the sleep, mood, and cognitive disturbances noted in the majority of patients with fibromyalgia (FM) [1,2,66]. Phasic alpha sleep activity is most characteristic of FM [67]. Some data suggest that disordered sleep patterns precede the development of pain and that abnormal sleep and pain predict depressive symptoms [66]. Some findings suggest a generalized hyperarousal state in FM. As an example, women with FM have similar nocturnal sleep disturbance to women with rheumatoid arthritis, but patients with FM report greater self-rated daytime sleepiness and fatigue [68].

A longitudinal study of 12,350 women in Norway who did not have musculoskeletal pain or physical impairments at baseline found incident FM in 327 women at follow-up [69]. There was a dose-dependent association between sleep problems and risk of FM, with an adjusted relative risk (RR) of FM of 3.43 (95% CI 2.26-5.19) among women who reported having sleep problems often or always, compared with women who never experienced sleep problems. Age-stratified analysis showed that women age ≥45 years who reported having sleep problems often or always had an adjusted RR of FM of 5.41 (95% CI 2.65-11.05), while the corresponding RR for women ages 20 to 44 years who reported having sleep problems often or always was 2.98 (95% CI 1.76-5.05).

In a prospective, population-based study, 19 percent of more than 4000 older adults (≥50 years of age) reported new widespread pain at follow-up. Nonrestorative sleep was the strongest independent predictor of new-onset widespread pain [70].

Sleep, mood, and cognitive disturbances are each interconnected and correlate with pain severity in FM, as noted in brain imaging studies discussed above [42-44]. Compared with patients with osteoarthritis and healthy controls, sleep quality was lowest and there was greater anxiety and depression in patients with FM [71]. However, there were no significant differences in polysomnographic measures of total sleep time, sleep latency, and total wake-after-sleep onset in the three groups. Furthermore, levels of alpha-delta sleep were statistically similar in FM and osteoarthritis. The quality of sleep was a strong mediator of attention, cognitive tests, and pain severity in FM [72].

STRESS/AUTONOMIC NERVOUS SYSTEM (ANS) DYSFUNCTION — The association of pain with sleep, mood, and cognitive abnormalities has been linked to stress reactivity and autonomic nervous system (ANS) dysfunction in fibromyalgia (FM). Sleep disturbances increase pain, which increases sympathetic cardiovascular reactivity [73].

Hypothalamic pituitary axis (HPA) and stress — Hyperactivity of the stress response, demonstrated by abnormalities of the hypothalamic-pituitary-adrenal (HPA) axis, has been found using different baseline and provocative testing, although the precise nature of these changes has not been elucidated [74]. There was a correlation between cerebrospinal levels of corticotropin-releasing factor, sensory pain, and variation in autonomic function in patients with FM [75]. There was also a strong correlation between cortisol levels and pain upon awakening and one hour after waking in patients with FM compared with controls [76]. Subjects with chronic widespread pain had higher serum cortisol levels than controls, and there was a significant correlation of HPA axis dysfunction with developing chronic widespread pain [77]. Serum cortisol levels, considered a proxy for stress, varied with the severity of neuropsychological deficits in patients with FM [78], and a systematic review failed to find a correlation of cortisol levels or reactivity with pain in patients with FM [79].

Evidence for ANS dysfunction include:

Decreased responsiveness to beta-adrenergic stimulation in those with FM was demonstrated by in-vitro testing of beta adrenergic receptor-mediated cyclic AMP generation [80].

In a study involving 58 women, including patients with FM and healthy age-matched controls, urinary catecholamines and heart rate were assessed for a 24-hour period in a controlled hospital setting (including relaxation, a test with prolonged mental stress, and sleep) and during daily activity [81].The catecholamine levels were lower in patients with FM than in controls. Patients with FM had significantly lower adrenaline levels during the night and the second day and had significantly lower dopamine levels during the first day, the night, and the second day. Overall, heart rate was significantly higher in patients than in controls.

In another approach, plasma catecholamines and ACTH were reduced in 16 patients with FM compared with 16 healthy controls as they performed static knee extension until exhaustion [82].

Nocturnal heart rate variability (HRV) indices were significantly different in women with FM compared with healthy individuals [81]. In patients with FM, these HRV parameters correlated with several symptoms including pain severity.

Nocturnal HRV indices indicative of sympathetic predominance were significantly different in women with FM when compared with healthy individuals [83]. In patients with FM, these HRV parameters correlated with several symptoms including pain severity. Opposite associations were seen in controls. They concluded that nocturnal HRV analyses are potential FM biomarkers. Patients with FM often demonstrate a hypertonic stress response, including increased blood pressure, when exposed to a painful stimulus [84].

Alterations in skin conductance, an indirect measure of sweating, were found in patients with FM compared with controls, and skin conductance varied less with stress and pain in FM [85].

PERIPHERAL PAIN MECHANISMS — Peripheral pain generators in fibromyalgia (FM) patients may include myofascial trigger points, ligamentous trigger points, or osteoarthritis of the joints and spine [86]. For example, FM is more common in patients with systemic rheumatic diseases or regional pain, such as chronic low back pain or complex regional pain syndrome. (See "Overview of chronic widespread (centralized) pain in the rheumatic diseases".)

Small fiber neuropathy – Several studies have suggested that there may be a relationship between FM and small fiber neuropathy (SFN) [87-90]. SFN has been defined by a skin biopsy demonstrating reduced intraepidermal nerve fiber (IENF) density in these reports.

A case-control study compared the function and morphology of small nerve fibers in 25 patients with FM syndrome with patients with depression and with healthy controls [87]. Patients with FM syndrome had increased levels of neuropathic pain based upon responses to questionnaires designed to assess this type of pain. Additionally, patients with FM syndrome but not patients with depression had impaired small fiber nerve function compared with controls, demonstrating increased cold- and warm-detection thresholds in quantitative sensory testing (QST), increased N1 latencies upon stimulation at the feet, and reduced amplitudes of pain-related evoked potentials upon stimulation of face, hands, and feet.

A study involving 27 patients with FM and 30 controls found that a significantly greater proportion of patients with FM had abnormal skin biopsies demonstrating findings consistent with small fiber peripheral neuropathy (41 versus 3 percent), suggesting that an underlying small-fiber polyneuropathy may cause symptoms sometimes identified as being due to FM [88]. Another uncontrolled study of patients referred for the evaluation of primary FM without associated medical comorbidities identified 6 of 20 patients who met criteria for small fiber neuropathy; electrodiagnostic studies were normal in all patients [89].

A study involving 41 patients with FM and 47 healthy controls reported findings suggestive of both a diffuse and a length-dependent neuropathic process [90]. There was evidence of stocking distribution hypesthesia in all patients with FM.

A systematic review of 222 patients with FM reported an estimated prevalence of 30 to 76 percent of SFN in FM, with a moderately high level of heterogeneity [91]. The authors concluded that 49 percent of patients with FM have "structural abnormalities of the small nerve fibers." One-hundred and fifty-five patients with FM with neuropathic symptoms underwent skin biopsies for SFN and nerve conduction studies [92]. Sixty percent were skin biopsy negative, 28 percent demonstrated distal extremity-reduced IENF density, and 12 percent had proximal extremity-reduced IENF density. Sural and medial plantar nerve conduction slowing correlated with reduced IENF density, as did markers of metabolic syndrome. However, pain quality and intensity did not distinguish patient subgroups.

However, there is a lack of consensus whether the association of reduced IENF with FM has any pathophysiologic significance [93]. For example, reduced IENF density has been found in conditions not typically associated with pain, such as amyotrophic lateral sclerosis [94]. SFN has been commonly noted in patients with complex, chronic pelvic pain [95], and there has been no correlation of reduced IENF density with neuropathic pain symptoms [96].

Clinically, SFN presents with a very different pain profile than FM. The finding of SFN in the lower extremities would not account for the chronic widespread pain involving the neck, shoulders, chest wall, and trunk or the chronic fatigue, mood and sleep disturbances characteristic of FM. Brain imaging in patients with SFN demonstrates structural and functional changes characteristic of central sensitization [97,98]. In a proof-of-concept animal study, increasing endogenous glutamate in the insula caused pain behavior and decrease in IENF density [99].

Although there has been no evidence for structural muscle abnormalities in FM [5,6], a few reports have noted metabolic changes in the muscle of patients with FM [100]. In one study, comparing 19 patients with FM and 14 controls, concentrations of adenosine triphosphate (ATP) and phosphocreatinine (PCr) were significantly lower (28 to 29 percent) in quadriceps muscle in the patients with FM [100]. The quadriceps muscle fat content was significantly greater in the patients with FM, who also exhibited lower physical capacity in the hands and legs, which correlated with the reduced concentrations of ATP and PCr. These findings were consistent with changes that could result from a combination of inactivity related to pain and muscle mitochondrial dysfunction.

One study has suggested that alterations in skeletal muscle, such as altered muscle fiber size distribution and decreased capillary density, may contribute to postexertional fatigue in FM [101]. In this study, patients with FM exhibited greater variability in muscle fiber size and altered fiber size distribution compared with controls. patients with FM with the highest percentage of type-1 muscle fibers recovered strength most effectively, and this was correlated with capillary density. However, overall, capillary density was lower in the patients with FM.

IMMUNE ABNORMALITIES — There is limited evidence to support the concept that fibromyalgia (FM) is an immune-mediated disorder [1]. A 2011 systematic review and meta-analysis concluded that the role of cytokines in FM is unclear [102]. A systematic analysis found significant differences in peripheral blood cytokine profiles in patients with FM compared with healthy controls, but there was no distinct pro- or anti-inflammatory pattern [103]. The secretion of interleukin 5 (IL-5) was associated with pain and specific monocyte subsets in patients with FM [104].

SUMMARY

There is conclusive evidence that alterations in central nervous system (CNS) pain processing are responsible for many of the features of fibromyalgia (FM). Brain imaging has provided the strongest evidence for this central sensitization, including an exaggerated pain response to experimental pain stimulus, altered structural and neurotransmitter function, and changes in resting-state functional connectivity. Studies suggest that an FM neurologic signature may be useful diagnostically and in future therapeutic trials [2]. (See 'Introduction' above and 'Terminology and pain classification' above and 'Central nervous system altered pain processing' above.)

Sleep, mood, and cognitive disturbances, as well as stress-related factors involving the autonomic nervous system (ANS), each contribute to the CNS hyperirritability. (See 'Sleep/mood/cognitive abnormalities' above.)

Genetic and environmental factors likely interact to promote a state of chronic central and peripheral nervous system hyperirritability. (See 'Genetic predisposition and candidate genes' above.)

Peripheral factors that may augment central pain processing include peripheral nerves and muscle. Small fiber neuropathy (SFN), defined by skin biopsy demonstrating reduced intraepidermal nerve fiber density, has been noted commonly in FM, although its causal role is controversial.

  1. Sluka KA, Clauw DJ. Neurobiology of fibromyalgia and chronic widespread pain. Neuroscience 2016; 338:114.
  2. López-Solà M, Woo CW, Pujol J, et al. Towards a neurophysiological signature for fibromyalgia. Pain 2017; 158:34.
  3. Nijs J, George SZ, Clauw DJ, et al. Central sensitization in chronic pain conditions: Latest discoveries and their potential for precision medicine. Lancet Rheumatol 2021; 3:e383.
  4. Staud R. Abnormal pain modulation in patients with spatially distributed chronic pain: fibromyalgia. Rheum Dis Clin North Am 2009; 35:263.
  5. Häkkinen A, Häkkinen K, Hannonen P, Alen M. Force production capacity and acute neuromuscular responses to fatiguing loading in women with fibromyalgia are not different from those of healthy women. J Rheumatol 2000; 27:1277.
  6. Lund E, Kendall SA, Janerot-Sjøberg B, Bengtsson A. Muscle metabolism in fibromyalgia studied by P-31 magnetic resonance spectroscopy during aerobic and anaerobic exercise. Scand J Rheumatol 2003; 32:138.
  7. Buskila D, Sarzi-Puttini P. Biology and therapy of fibromyalgia. Genetic aspects of fibromyalgia syndrome. Arthritis Res Ther 2006; 8:218.
  8. Arnold LM, Hudson JI, Hess EV, et al. Family study of fibromyalgia. Arthritis Rheum 2004; 50:944.
  9. Limer KL, Nicholl BI, Thomson W, McBeth J. Exploring the genetic susceptibility of chronic widespread pain: the tender points in genetic association studies. Rheumatology (Oxford) 2008; 47:572.
  10. Smith SB, Maixner DW, Fillingim RB, et al. Large candidate gene association study reveals genetic risk factors and therapeutic targets for fibromyalgia. Arthritis Rheum 2012; 64:584.
  11. Arnold LM, Fan J, Russell IJ, et al. The fibromyalgia family study: A genome-wide linkage scan study. Arthritis Rheum 2013; 65:1122.
  12. Jones KD, Gelbart T, Whisenant TC, et al. Genome-wide expression profiling in the peripheral blood of patients with fibromyalgia. Clin Exp Rheumatol 2016; 34:S89.
  13. Rahman MS, Winsvold BS, Chavez Chavez SO, et al. Genome-wide association study identifies RNF123 locus as associated with chronic widespread musculoskeletal pain. Ann Rheum Dis 2021; 80:1227.
  14. Martínez-Jauand M, Sitges C, Rodríguez V, et al. Pain sensitivity in fibromyalgia is associated with catechol-O-methyltransferase (COMT) gene. Eur J Pain 2013; 17:16.
  15. Inanir A, Karakus N, Ates O, et al. Clinical symptoms in fibromyalgia are associated to catechol-O-methyltransferase (COMT) gene Val158Met polymorphism. Xenobiotica 2014; 44:952.
  16. Park DJ, Kim SH, Nah SS, et al. Association between catechol-O-methyl transferase gene polymorphisms and fibromyalgia in a Korean population: A case-control study. Eur J Pain 2016; 20:1131.
  17. Lee C, Liptan G, Kantorovich S, et al. Association of Catechol-O-methyltransferase single nucleotide polymorphisms, ethnicity, and sex in a large cohort of fibromyalgia patients. BMC Rheumatol 2018; 2:38.
  18. Nicholl BI, Holliday KL, Macfarlane GJ, et al. Association of HTR2A polymorphisms with chronic widespread pain and the extent of musculoskeletal pain: results from two population-based cohorts. Arthritis Rheum 2011; 63:810.
  19. Tanwar S, Mattoo B, Kumar U, et al. Does human serotonin-1A receptor polymorphism (rs6295) code for pain and associated symptoms in fibromyalgia syndrome? Reumatismo 2021; 73:24.
  20. Solak Ö, Erdoğan MÖ, Yıldız H, et al. Assessment of opioid receptor μ1 gene A118G polymorphism and its association with pain intensity in patients with fibromyalgia. Rheumatol Int 2014; 34:1257.
  21. Kosek E, Martinsen S, Gerdle B, et al. The translocator protein gene is associated with symptom severity and cerebral pain processing in fibromyalgia. Brain Behav Immun 2016; 58:218.
  22. Tour J, Löfgren M, Mannerkorpi K, et al. Gene-to-gene interactions regulate endogenous pain modulation in fibromyalgia patients and healthy controls-antagonistic effects between opioid and serotonin-related genes. Pain 2017; 158:1194.
  23. Ellerbrock I, Sandström A, Tour J, et al. Polymorphisms of the μ-opioid receptor gene influence cerebral pain processing in fibromyalgia. Eur J Pain 2021; 25:398.
  24. Estévez-López F, Camiletti-Moirón D, Aparicio VA, et al. Identification of candidate genes associated with fibromyalgia susceptibility in southern Spanish women: the al-Ándalus project. J Transl Med 2018; 16:43.
  25. Park DJ, Kim SH, Nah SS, et al. Association between brain-derived neurotrophic factor gene polymorphisms and fibromyalgia in a Korean population: a multicenter study. Arthritis Res Ther 2018; 20:220.
  26. Feng J, Zhang Z, Wu X, et al. Discovery of potential new gene variants and inflammatory cytokine associations with fibromyalgia syndrome by whole exome sequencing. PLoS One 2013; 8:e65033.
  27. Ciampi de Andrade D, Maschietto M, Galhardoni R, et al. Epigenetics insights into chronic pain: DNA hypomethylation in fibromyalgia-a controlled pilot-study. Pain 2017; 158:1473.
  28. Polli A, Ghosh M, Bakusic J, et al. DNA Methylation and Brain-Derived Neurotrophic Factor Expression Account for Symptoms and Widespread Hyperalgesia in Patients With Chronic Fatigue Syndrome and Comorbid Fibromyalgia. Arthritis Rheumatol 2020; 72:1936.
  29. Dutta D, Brummett CM, Moser SE, et al. Heritability of the Fibromyalgia Phenotype Varies by Age. Arthritis Rheumatol 2020; 72:815.
  30. O'Brien AT, Deitos A, Triñanes Pego Y, et al. Defective Endogenous Pain Modulation in Fibromyalgia: A Meta-Analysis of Temporal Summation and Conditioned Pain Modulation Paradigms. J Pain 2018; 19:819.
  31. Jensen KB, Kosek E, Petzke F, et al. Evidence of dysfunctional pain inhibition in Fibromyalgia reflected in rACC during provoked pain. Pain 2009; 144:95.
  32. Julien N, Goffaux P, Arsenault P, Marchand S. Widespread pain in fibromyalgia is related to a deficit of endogenous pain inhibition. Pain 2005; 114:295.
  33. Montoya P, Sitges C, García-Herrera M, et al. Reduced brain habituation to somatosensory stimulation in patients with fibromyalgia. Arthritis Rheum 2006; 54:1995.
  34. Salemi S, Aeschlimann A, Wollina U, et al. Up-regulation of delta-opioid receptors and kappa-opioid receptors in the skin of fibromyalgia patients. Arthritis Rheum 2007; 56:2464.
  35. Harris RE, Clauw DJ, Scott DJ, et al. Decreased central mu-opioid receptor availability in fibromyalgia. J Neurosci 2007; 27:10000.
  36. Russell IJ, Orr MD, Littman B, et al. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum 1994; 37:1593.
  37. Haas L, Portela LV, Böhmer AE, et al. Increased plasma levels of brain derived neurotrophic factor (BDNF) in patients with fibromyalgia. Neurochem Res 2010; 35:830.
  38. Hubbard CS, Lazaridou A, Cahalan CM, et al. Aberrant Salience? Brain Hyperactivation in Response to Pain Onset and Offset in Fibromyalgia. Arthritis Rheumatol 2020; 72:1203.
  39. Staud R, Godfrey MM, Robinson ME. Fibromyalgia Patients Are Not Only Hypersensitive to Painful Stimuli But Also to Acoustic Stimuli. J Pain 2021; 22:914.
  40. Gracely RH, Petzke F, Wolf JM, Clauw DJ. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum 2002; 46:1333.
  41. Napadow V, Harris RE. What has functional connectivity and chemical neuroimaging in fibromyalgia taught us about the mechanisms and management of 'centralized' pain? Arthritis Res Ther 2014; 16:425.
  42. Giesecke T, Gracely RH, Williams DA, et al. The relationship between depression, clinical pain, and experimental pain in a chronic pain cohort. Arthritis Rheum 2005; 52:1577.
  43. Loggia ML, Berna C, Kim J, et al. Disrupted brain circuitry for pain-related reward/punishment in fibromyalgia. Arthritis Rheumatol 2014; 66:203.
  44. Kamping S, Bomba IC, Kanske P, et al. Deficient modulation of pain by a positive emotional context in fibromyalgia patients. Pain 2013; 154:1846.
  45. Lee J, Protsenko E, Lazaridou A, et al. Encoding of Self-Referential Pain Catastrophizing in the Posterior Cingulate Cortex in Fibromyalgia. Arthritis Rheumatol 2018; 70:1308.
  46. Müller M, Wüthrich F, Federspiel A, et al. Altered central pain processing in fibromyalgia-A multimodal neuroimaging case-control study using arterial spin labelling. PLoS One 2021; 16:e0235879.
  47. Kuchinad A, Schweinhardt P, Seminowicz DA, et al. Accelerated brain gray matter loss in fibromyalgia patients: premature aging of the brain? J Neurosci 2007; 27:4004.
  48. Burgmer M, Gaubitz M, Konrad C, et al. Decreased gray matter volumes in the cingulo-frontal cortex and the amygdala in patients with fibromyalgia. Psychosom Med 2009; 71:566.
  49. Hsu MC, Harris RE, Sundgren PC, et al. No consistent difference in gray matter volume between individuals with fibromyalgia and age-matched healthy subjects when controlling for affective disorder. Pain 2009; 143:262.
  50. Pomares FB, Funck T, Feier NA, et al. Histological Underpinnings of Grey Matter Changes in Fibromyalgia Investigated Using Multimodal Brain Imaging. J Neurosci 2017; 37:1090.
  51. Jensen KB, Srinivasan P, Spaeth R, et al. Overlapping structural and functional brain changes in patients with long-term exposure to fibromyalgia pain. Arthritis Rheum 2013; 65:3293.
  52. Kim DJ, Lim M, Kim JS, et al. Altered white matter integrity in the corpus callosum in fibromyalgia patients identified by tract-based spatial statistical analysis. Arthritis Rheumatol 2014; 66:3190.
  53. Cagnie B, Coppieters I, Denecker S, et al. Central sensitization in fibromyalgia? A systematic review on structural and functional brain MRI. Semin Arthritis Rheum 2014; 44:68.
  54. Harris RE, Sundgren PC, Craig AD, et al. Elevated insular glutamate in fibromyalgia is associated with experimental pain. Arthritis Rheum 2009; 60:3146.
  55. Wood PB, Schweinhardt P, Jaeger E, et al. Fibromyalgia patients show an abnormal dopamine response to pain. Eur J Neurosci 2007; 25:3576.
  56. Mhalla A, de Andrade DC, Baudic S, et al. Alteration of cortical excitability in patients with fibromyalgia. Pain 2010; 149:495.
  57. Valdés M, Collado A, Bargalló N, et al. Increased glutamate/glutamine compounds in the brains of patients with fibromyalgia: a magnetic resonance spectroscopy study. Arthritis Rheum 2010; 62:1829.
  58. Foerster BR, Petrou M, Edden RA, et al. Reduced insular γ-aminobutyric acid in fibromyalgia. Arthritis Rheum 2012; 64:579.
  59. Harper DE, Ichesco E, Schrepf A, et al. Resting Functional Connectivity of the Periaqueductal Gray Is Associated With Normal Inhibition and Pathological Facilitation in Conditioned Pain Modulation. J Pain 2018; 19:635.e1.
  60. Truini A, Tinelli E, Gerardi MC, et al. Abnormal resting state functional connectivity of the periaqueductal grey in patients with fibromyalgia. Clin Exp Rheumatol 2016; 34:S129.
  61. Fallon N, Chiu Y, Nurmikko T, Stancak A. Functional Connectivity with the Default Mode Network Is Altered in Fibromyalgia Patients. PLoS One 2016; 11:e0159198.
  62. Martucci KT, Weber KA 2nd, Mackey SC. Altered Cervical Spinal Cord Resting-State Activity in Fibromyalgia. Arthritis Rheumatol 2019; 71:441.
  63. Kaplan CM, Schrepf A, Vatansever D, et al. Functional and neurochemical disruptions of brain hub topology in chronic pain. Pain 2019; 160:973.
  64. Larkin TE, Kaplan CM, Schrepf A, et al. Altered network architecture of functional brain communities in chronic nociplastic pain. Neuroimage 2021; 226:117504.
  65. Hsiao FJ, Chen WT, Ko YC, et al. Neuromagnetic Amygdala Response to Pain-Related Fear as a Brain Signature of Fibromyalgia. Pain Ther 2020; 9:765.
  66. Moldofsky H. The significance of dysfunctions of the sleeping/waking brain to the pathogenesis and treatment of fibromyalgia syndrome. Rheum Dis Clin North Am 2009; 35:275.
  67. Roizenblatt S, Neto NS, Tufik S. Sleep disorders and fibromyalgia. Curr Pain Headache Rep 2011; 15:347.
  68. Roehrs T, Diederichs C, Gillis M, et al. Nocturnal sleep, daytime sleepiness and fatigue in fibromyalgia patients compared to rheumatoid arthritis patients and healthy controls: a preliminary study. Sleep Med 2013; 14:109.
  69. Mork PJ, Nilsen TI. Sleep problems and risk of fibromyalgia: longitudinal data on an adult female population in Norway. Arthritis Rheum 2012; 64:281.
  70. McBeth J, Lacey RJ, Wilkie R. Predictors of new-onset widespread pain in older adults: results from a population-based prospective cohort study in the UK. Arthritis Rheumatol 2014; 66:757.
  71. Yeung WK, Morgan K, Mckenna F. Comparison of sleep structure and psychometric profiles in patients with fibromyalgia, osteoarthritis and healthy controls. J Sleep Res 2018; 27:290.
  72. Fang SC, Wu YL, Chen SC, et al. Subjective sleep quality as a mediator in the relationship between pain severity and sustained attention performance in patients with fibromyalgia. J Sleep Res 2019; 28:e12843.
  73. Rizzi M, Radovanovic D, Santus P, et al. Influence of autonomic nervous system dysfunction in the genesis of sleep disorders in fibromyalgia patients. Clin Exp Rheumatol 2017; 35 Suppl 105:74.
  74. Adler GK, Kinsley BT, Hurwitz S, et al. Reduced hypothalamic-pituitary and sympathoadrenal responses to hypoglycemia in women with fibromyalgia syndrome. Am J Med 1999; 106:534.
  75. McLean SA, Williams DA, Stein PK, et al. Cerebrospinal fluid corticotropin-releasing factor concentration is associated with pain but not fatigue symptoms in patients with fibromyalgia. Neuropsychopharmacology 2006; 31:2776.
  76. McLean SA, Williams DA, Harris RE, et al. Momentary relationship between cortisol secretion and symptoms in patients with fibromyalgia. Arthritis Rheum 2005; 52:3660.
  77. McBeth J, Silman AJ, Gupta A, et al. Moderation of psychosocial risk factors through dysfunction of the hypothalamic-pituitary-adrenal stress axis in the onset of chronic widespread musculoskeletal pain: findings of a population-based prospective cohort study. Arthritis Rheum 2007; 56:360.
  78. Barceló-Martinez E, Gelves-Ospina M, Navarro Lechuga E, et al. Serum cortisol levels and neuropsychological impairments in patients diagnosed with Fibromyalgia. Actas Esp Psiquiatr 2018; 46:1.
  79. Úbeda-D'Ocasar E, Jiménez Díaz-Benito V, Gallego-Sendarrubias GM, et al. Pain and Cortisol in Patients with Fibromyalgia: Systematic Review and Meta-Analysis. Diagnostics (Basel) 2020; 10.
  80. Maekawa K, Twe C, Lotaif A, et al. Function of beta-adrenergic receptors on mononuclear cells in female patients with fibromyalgia. J Rheumatol 2003; 30:364.
  81. Riva R, Mork PJ, Westgaard RH, et al. Catecholamines and heart rate in female fibromyalgia patients. J Psychosom Res 2012; 72:51.
  82. Kadetoff D, Kosek E. Evidence of reduced sympatho-adrenal and hypothalamic-pituitary activity during static muscular work in patients with fibromyalgia. J Rehabil Med 2010; 42:765.
  83. Lerma C, Martinez A, Ruiz N, et al. Nocturnal heart rate variability parameters as potential fibromyalgia biomarker: correlation with symptoms severity. Arthritis Res Ther 2011; 13:R185.
  84. Reyes del Paso GA, Duschek S. Responses to a comment on "autonomic cardiovascular control and responses to experimental pain stimulation in fibromyalgia syndrome". J Psychosom Res 2012; 72:87.
  85. Reyes Del Paso GA, de la Coba P. Reduced activity, reactivity and functionality of the sympathetic nervous system in fibromyalgia: An electrodermal study. PLoS One 2020; 15:e0241154.
  86. Staud R. Peripheral pain mechanisms in chronic widespread pain. Best Pract Res Clin Rheumatol 2011; 25:155.
  87. Üçeyler N, Zeller D, Kahn AK, et al. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013; 136:1857.
  88. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain 2013; 154:2310.
  89. Giannoccaro MP, Donadio V, Incensi A, et al. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve 2014; 49:757.
  90. Caro XJ, Winter EF. Evidence of abnormal epidermal nerve fiber density in fibromyalgia: clinical and immunologic implications. Arthritis Rheumatol 2014; 66:1945.
  91. Grayston R, Czanner G, Elhadd K, et al. A systematic review and meta-analysis of the prevalence of small fiber pathology in fibromyalgia: Implications for a new paradigm in fibromyalgia etiopathogenesis. Semin Arthritis Rheum 2019; 48:933.
  92. Lawson VH, Grewal J, Hackshaw KV, et al. Fibromyalgia syndrome and small fiber, early or mild sensory polyneuropathy. Muscle Nerve 2018; 58:625.
  93. Clauw DJ. What is the meaning of "small fiber neuropathy" in fibromyalgia? Pain 2015; 156:2115.
  94. Dalla Bella E, Lombardi R, Porretta-Serapiglia C, et al. Amyotrophic lateral sclerosis causes small fiber pathology. Eur J Neurol 2016; 23:416.
  95. Chen A, De E, Argoff C. Small Fiber Polyneuropathy Is Prevalent in Patients Experiencing Complex Chronic Pelvic Pain. Pain Med 2019; 20:521.
  96. Gandhi RA, Selvarajah D. Understanding and treating painful diabetic neuropathy: time for a paradigm shift. Diabet Med 2015; 32:771.
  97. Hsieh PC, Tseng MT, Chao CC, et al. Imaging signatures of altered brain responses in small-fiber neuropathy: reduced functional connectivity of the limbic system after peripheral nerve degeneration. Pain 2015; 156:904.
  98. Chao CC, Tseng MT, Lin YH, et al. Brain imaging signature of neuropathic pain phenotypes in small-fiber neuropathy: altered thalamic connectome and its associations with skin nerve degeneration. Pain 2021; 162:1387.
  99. Harte SE, Clauw DJ, Hayes JM, et al. Reduced intraepidermal nerve fiber density after a sustained increase in insular glutamate: a proof-of-concept study examining the pathogenesis of small fiber pathology in fibromyalgia. Pain Rep 2017; 2:e590.
  100. Gerdle B, Forsgren MF, Bengtsson A, et al. Decreased muscle concentrations of ATP and PCR in the quadriceps muscle of fibromyalgia patients--a 31P-MRS study. Eur J Pain 2013; 17:1205.
  101. Srikuea R, Symons TB, Long DE, et al. Association of fibromyalgia with altered skeletal muscle characteristics which may contribute to postexertional fatigue in postmenopausal women. Arthritis Rheum 2013; 65:519.
  102. Uçeyler N, Häuser W, Sommer C. Systematic review with meta-analysis: cytokines in fibromyalgia syndrome. BMC Musculoskelet Disord 2011; 12:245.
  103. O'Mahony LF, Srivastava A, Mehta P, Ciurtin C. Is fibromyalgia associated with a unique cytokine profile? A systematic review and meta-analysis. Rheumatology (Oxford) 2021; 60:2602.
  104. Merriwether EN, Agalave NM, Dailey DL, et al. IL-5 mediates monocyte phenotype and pain outcomes in fibromyalgia. Pain 2021; 162:1468.
Topic 5626 Version 18.0

References