You are on page 1of 12

ARTHRITIS & RHEUMATISM

Vol. 40, No. 11, November 1997, pp 1928-1939


1928 0 1997. American College of Rheumatology

CONFERENCE SUMMARY

THE NEUROSCIENCE AND ENDOCRINOLOGY OF FIBROMYALGIA

STANLEY R. PILLEMER, LAURENCE A. BRADLEY, LESLIE J. CROFFORD,


HARVEY MOLDOFSKY, and GEORGE P. CHROUSOS

Fibromyalgia syndrome (FMS) is characterized Chronic pain segment


by widespread pain and tenderness in reproducible,
Pain mechanisms and physiology. Pain percep-
characteristic anatomic locations (1). The syndrome is
tion initiated by a noxious peripheral stimulus is medi-
associated with chronic pain and dysregulation of neu-
ated by ascending neural pathways, as shown in Figure
roendocrine function and of sleep. In addition, patients
1A (2). However, the transmission of nociceptive signals
often experience high levels of fatigue and other symp-
from peripheral tissues to the brain is modulated by
toms, such as irritable bowel or bladder.
endogenous and exogenous processes. For example,
Recently, researchers in fields other than the
several descending pain inhibitory fibers originate in
rheumatic diseases have made considerable progress in
brain stem sites and inhibit spinal nociceptive transmis-
studying the individual component manifestations of
sion (Figure 1B). In addition, various psychological,
FMS. Therefore, several experts in the fields of pain,
pharmacologic, and physical interventions can attenuate
neuroendocrine function, and sleep research were in-
or promote nociceptive processing and thus affect pain
vited to meet with a small group of expert FMS investi-
perception. These modulating effects may be mediated
gators at an interdisciplinary workshop held July 16-17,
by the activation or suppression of a complex network of
1996 at the National Institutes of Health, Bethesda,
cerebral, bulbar, and spinal neurons. For example, elec-
Maryland. The major goals of the meeting were to
trical stimulation or destruction of central nervous sys-
heighten awareness among rheumatologists regarding
advances in clinical and basic research in chronic pain, tem (CNS) structures and pathways can produce pain or
neuroendocrine function, and sleep, and, conversely, to analgesia. In addition, physical trauma and diseases that
inform investigators in these research areas about FMS. directly affect the CNS, such as stroke or multiple
This report summarizes the material presented at the sclerosis, can produce central pain syndromes. Investi-
workshop, which was organized into 3 segments corre- gators are beginning to examine the mechanisms and
sponding to the research areas noted above. physiology of pain with new imaging methods for study-
ing nociceptive processing in animals and in the con-
scious human brain. These methods, which include
Cosponsors of the Workshop on the Neuroscience and En- positron emission tomography (PET), single-photon
docrinology of Fibromyalgia: National Institute of Arthritis and Mus- emission computed tomography (SPECT), and func-
culoskeletal and Skin Diseases; National Institute of Nursing Re-
search; Office of Behavioral and Social Sciences Research; Office of tional magnetic resonance imaging, may clarify neuro-
Research on Women’s Health; and National Center on Sleep Disor- logic processes involved in chronic pain disorders such as
ders Research, National Heart, Lung, and Blood Institute. FMS (3,4).
Stanley R. Pillemer, MD: National Institute of Arthritis and
Musculoskeletal and Skin Diseases, National Institutes of Health, Spinal cord and brain plasticity. Peripheral tis-
Bethesda, Maryland; Laurence A. Bradley, PhD: The University of sue or nerve injury often results in hyperalgesia, in which
Alabama at Birmingham; Leslie J. Crofford, MD: University of
Michigan, Ann Arbor; Harvey Moldofsky, MD: Toronto Western innocuous stimuli such as light touch may be perceived
Hospital, Toronto, Ontario, Canada; George P. Chrousos, MD, PhD: as painful. Hyperalgesia is related to changes at the site
National Institute of Child Health and Human Development, National of injury as well as to CNS hyperexcitability that leads to
Institutes of Health, Bethesda, Maryland.
Address reprint requests to Stanley R. Pillemer, MD, Office long-term changes in the nervous system, referred to as
of the Director, National Institute of Arthritis and Musculoskeletal plasticity (5-7).
and Skin Diseases, Building 45, Room 5As-37G, 45 Center Drive MSC Animal models of hyperalgesia produced by in-
6500, Bethesda, MD 20892-6500.
Submitted for publication March 26,1997; accepted in revised flammation or nerve injury mimic persistent pain condi-
form June 10, 1997. tions. These models suggest that, after nerve injury, new
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1929

r-7
Limbic forebrain
Frontal lobe
Limbic forebrain

Thalamus/ Thalamus/
Hypothalamus Hypothalamus

Sl/S2/ lnsula
Cortex \ Cortex

Midbrain Midbrain

Medulla

Spinal cord
-
(C3 C4)

Figure 1. A, Ascending neural pathways for pain. Pain perception is normally initiated by activation of
nociceptors in peripheral tissues. Primary nociceptive afferents ascend contralaterally and excite spinothalamic
and spinoreticular neurons in the dorsal horn of the spinal cord through neurotransmitters such as glutamate,
aspartate, substance P, and calcitonin gene-related peptide. The signals generated in these neurons are then
transmitted to the brain via their axons, which travel in 3 primary ascending tracts that project to the thalamus
and the reticular formation. Direct spinothalamic tract projections to the ventral posterior lateral thalamus excite
neurons with connection to the primary (Sl) and secondary (S2) somatosensory and insular cortex. The S l cortex
mediates the discrimination of intensity and spatial and temporal aspects of pain. The limbic system is also
involved in nociceptive input. The above pathways are also interconnected with neurons involved in mediating
autonomic, endocrine, mnemonic, and hedonic components. B, Descending pathways for pain inhibition.
Corticothalamic neurons from the S1 and S2 somatosensory cortex modulate the excitability of neurons in the
ventral posterior lateral thalamus. Frontal lobe and limbic forebrain projections activate medial thalamic,
hypothalamic, and brain stem reticular formation neurons, which modulate the spinal (and trigeminal) dorsal
horn through the dorsolateral spinal cord. (Adapted, with permission of the American Academy of Neurology,
from Casey KL, Gonzales GR, Max MB, Price DD, Portenoy RK, Rowbotham MC: Continuum; Pain 2, 1996.)

axon sprouts that are sensitive to stimulation often mechanical, thermal, and chemical stimuli (Figure 2).
develop in the injured zone. Spontaneous discharges This hyperexcitability of CNS neurons, called central
from these damaged nerves, as well as from the dorsal sensitization, leads to increased activity at higher brain
root ganglion, increase the neuronal barrage into the centers. It is likely perceived as more intense and
CNS and contribute to increased pain after injury. prolonged pain. The hyperexcitability is mediated by the
The responses of CNS neurons also change after activation of neurons with N-methyl-D-aspartate
injury. After inflammation, spinal dorsal horn nocicep- (NMDA) receptor sites by excitatory amino acids. The
tive neurons often exhibit an increased excitability that is release of these amino acids from presynaptic sites and
characterized by an enlargement of their peripheral their effects on dorsal horn neurons are enhanced by
receptive fields and an increased responsiveness to neuropeptide transmitters such as dynorphin, substance
1930 PILLEMER ET AL

Tissue or Nerve Injury


stimulation of tender and control points (9), indicating
greater sensitivity to this stimulation. In addition, pa-
tients with FMS tend to meet diagnostic criteria for
multiple lifetime psychiatric diagnoses, especially major
Excitatory amino acid release depression, whereas nonpatients do not differ from
neuropeptide release
healthy controls in psychiatric morbidity. Thus, persons
with FMS, regardless of their behavior in seeking health
care and their psychiatric histories, differ from healthy
persons in neurosensory processing of stimulation at
numerous anatomic sites.
and Hyperexcitability Substance P, regional cerebral blood flow, and
Loss of Inhibition
FMS pain. The behavioral evidence of altered CNS
Increased Pain processing of nociceptive stimuli in persons with FMS is
supported by evidence of biologic abnormalities. For
Figure 2. Changes in the nervous syatem after injury. The hyperexcit-
ability involves activation of neurons at N-methyl-u-aspartate
example, SP, which is an important nociceptive neuro-
(NMDA) receptor sites by excitatory amino acids. The release of transmitter at the dorsal horn level, is elevated in the
excitatory amino acids from prcsynaptic sites and their effects on cerebrospinal fluid (CSF) of both patients and nonpa-
dorsal horn neurons are enhanced by neuropeptide transmitters such tients with FMS (10,ll). The high CSF levels of SP are
as dynorphin, substance P (SP), and calcitonin gene-related peptide stable and are associated with high CSF concentrations
(CGRP).
of nerve growth factor. Certain anti-nociceptive peptide
mediator levels are also abnormal in the CSF of FMS
patients. Met-enkephalin-arg-phe levels are low, while
P (SP), and calcitonin gene-related peptide. However, the N-terminal peptide fragment of SP (SP,-,) is in-
the dorsal horn neuron hyperexcitability and behavioral creased 2-fold (12). There is also indirect evidence of a
hyperalgesia may be attenuated in a dose-dependent decrease in the availability of serotonin to down-
manner by selectively acting NMDA receptor antag- regulate nociception in FMS (12).
onists. Finally, both patients and nonpatients with FMS
Descending projections from brain stem sites are characterized by significantly lower levels of regional
dampen or counteract the spinal cord hyperexcitability cerebral blood flow (rCBF) in the caudate nucleus, as
produced by tissue or nerve injury (Figure 1B). The net compared with controls (1 1). Low thalamic rCBF also
descending effects are greater after inflammation than has been observed in FMS patients, as well as in patients
in the absence of inflammation. with other disorders associated with chronic pain, such
These findings have important implications re- as chronic neuropathic pain, metastatic cancer, and
garding treatment, because the pain of tissue or nerve mononeuropathy (13-17). The caudatc nucleus and
damage can be attacked at the site of the injury where it thalamus are both involved in signaling the occurrence
is initiated, as well as at CNS sites where it is maintained. of noxious events. It may be that low rCBF, which
New treatment strategies for controlling persistent pain indicates decreased functional activity, is a marker for
following injury have been developed based on these impaired inhibition of nociceptive transmission by these
new research advances. brain structures. The findings reviewed above suggest
Fibromyalgia as a model for chronic pain. There that chronic pain in FMS is associated with several
is increasing behavioral and biologic evidence that, behavioral and biologic abnormalities indicative of al-
similar to nerve injury pain, the pathogenesis of FMS tered CNS processing of nociceptive signals.
pain is related to altered CNS processing of nociceptive Animal models of sex differences in pain and
stimuli. For example, the pain thresholds of FMS pa- analgesia. The high prevalence of chronic pain condi-
tients and nonpatients (individuals who meet critcria for tions, such as FMS, in women compared with men may
FMS but who do not seek medical treatment) in re- be related to sex differences in pain sensitivity. Studies in
sponse to pressure stimulation at diagnostic tender both animals and humans indicate that males show
points (1) and anatomic control sites are 2-3 times lower higher pain thresholds and tolerance when exposed to
than those of healthy persons (8,9). Patients and nonpa- noxious stimuli ( 17-21). Furthermore, females discrim-
tients also have significantly higher scores than controls inate better among painful stimuli than males, indicating
on an index of sensory discrimination ability during a higher sensitivity to these stimuli (17).
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1931

Animals also exhibit profound sex differences in needed to determine whether CBT protocols can reducc
the magnitude and mechanisms of analgesia produced maladaptive cognitions, decrcase pain and utilization of
by pharmacologic (administration of opioid agonists), health care, and enhance psychological and physical
electrical (brain stimulation-produced analgesia), or en- function in persons with FMS.
vironmental (stress exposure) manipulations (22). For Pain studies: methodologic issues. The evalua-
example, stress-induced analgesia (SIA) often occurs at tion of pain and pain mechanisms in FMS is problem-
higher levels in male rodents than in females (19). SIA atic. Tenderness at each anatomic site is usually mea-
may be mediated by opioid or non-opioid neurotrans- sured by determining the pain threshold to a single
mitters. As determined by sensitivity to specific receptor pressure stimulus of increasing intensity. This method
antagonists, non-opioid SIA in male mice appears to be may be influenced by factors such as variation in rate of
mediated by the NMDA subtype of excitatory amino increases in pressure; thus, results are easily biased.
acid receptors. In contrast, female mice display equipo- Moreover, the evaluation of tenderness in patients with
tent non-opioid SIA that is dependent on estrogen for its generalized body pain lacks the power of comparison
expression and is not sensitive to NMDA antagonist with a contralateral or baseline control. Dynamic designs
(22). The presence of such qualitative sex differences in that include evaluation of physiologic response3 during
brain neurochemistry related to analgesia processes baseline and stimulation conditions may be useful both
indicates possible sex differences in the clinical efficacy to increase experimental power and to evaluate mean-
of analgesic agents that may activate these mechanisms. ingful disease factors; for example, measurement of
Cognitive processes and the pain experience. brain rCBF by PET or SPECT during both baseline and
Patients with rheumatic diseases such as FMS vary in ipsilateral exposure to a controlled stimulus may de-
their responses to persistent pain. Three important crease measurement problems.
cognitive variables may explain how FMS patients adjust
to persistent pain. First, a high level of self-efficacy, or
Neuroendocrine function segment
the belief in one’s ability to engage in actions sufficient
to attain a desired health outcome, is associated with Neuroendocrine alterations associated with al-
reports of lower pain and disability among patients with tered sleep, mood, and pain perception. FMS is associ-
arthritis ( 2 3 ) . Increases in self-efficacy following coping ated with alterations in pain perception, mood, and
skills training interventions have been associated with sleep, and may be a “stress-related illness” since it., omet
long-term improvements in pain and function. Second, often coincides with physical or psychological stress.
patients who rate their ability to control and decrease During stress, CNS and peripheral functions generally
pain as high and who avoid the use of catastrophizing are altered to reestablish homeostasis and to preserve
coping strategies report low levels of pain and disability. life. The physiology of stress is complex and is subserved
Third, frequent use of adaptive, cognitive coping strate- by a dedicated stress system (28,29). This consists of
gies is also associated with lower levels of pain and CNS and peripheral components and includes the
disability. hypothalamic-pituitary-adrenal (HPA) axis and the au-
Newly developed treatment protocols for rheu- tonomic (sympathetic) system (Figure 3). In the brain,
matic disease patients use cognitive-behavioral inter- corticotropin-releasing hormone (CRH) neurons are
ventions such as cognitive restructuring and self- located in the paraventricular nucleus of the hypothala-
instructional training to increase self-efficacy beliefs and mus, the brain stem, the amygdala, and other sites. The
to reduce overly negative and maladaptive cognitions. sympathetic system centers include the locus ceruleus,
These protocols have been shown to be effective for which regulates arousal, and other brain stem nuclei
patients with rheumatoid arthritis and osteoarthritis in responsible for regulation of central and peripheral
randomized, controlled clinical trials (24,25). Recent sympathetic functions.
studies of cognitive-behavioral therapy (CBT) protocols The principal regulator of the HPA axis, CRH, is
have shown positive findings in patients with FMS (26). secreted into the hypophyseal portal system, where it
One randomized, controlled trial of a CBT protocol increases adrenocorticotropic hormone (ACTH) secre-
showed no clinical or economic benefits of treatment for tion, which in turn stimulates cortisol secretion by the
FMS (27); however, these findings appear to have been adrenal cortex. Cortisol then feeds back negatively to the
associated with poor patient adherence to the study pituitary, the hypothdamus, and higher centers of the
protocol. Thus, the effects of CBT for patients with FMS CNS that control HPA axis activity. The systemic sym-
have not been established. Controlled research is pathetic system primarily secretes norepinephrine, while
1932 PILLEMER ET AL

Circadian the opioid peptides inhibit both systems. Pro-


opiomelanocortin-derived peptides, such as a-
melanocyte-stimulating hormone and p-endorphin, in-
hibit both the HPA axis and the sympathetic system and
may be involved in peripheral stress-induced analgesia.
~ BEHAVIORAL ADAPTATION (& Interestingly, while SP is a potent inhibitor of CRH, it
stimulates the central sympathetic system. In addition,
_- * - GABAJBZD ----__- the HPA axis inhibits the reproductive axis, growth, and
the immune system.
In the classic stress response, CRH and arginine
vasopressin (AVP) work synergistically to cause ACTH
secretion. Neurons secrete CRH and AVP with 2-3
pulses per hour. Approximately 75% of these pulses are
secreted together, producing a synergistic effect on
ACTH secretion.
Pathophysiologic manifestations tend to occur
when the stress response is chronically hyperactive or
hypoactive. Individuals with melancholic depression
have a chronically hyperactive and hyperresponsive
\ ACTH I stress system. In contrast, there are disorders in which a

1
hyporesponsive stress system is noted. These include
seasonal affective disorder, chronic fatigue syndrome,
postpartum depression, and FMS (30-33). An under-
Gluc:oz&oids Epiriephrine standing of the neurotransmitters and neuropeptides
Norepinephrine that are stimulatory or inhibitory to the stress system
may suggest possible candidate genes for defining the
PERIPHERALADAPTATION genetic predisposition to hyperactivity or hypoactivity of
this system. SP, a potent inhibitor of CRH, may partic-
Figure 3. Interactions between the 2 major components of the stress
system, the hypothalamic-pituitary-adrenal axis system (left) and the ipate in causing the low activity of the CRH neuron
locus ceruleusinorepinephrine sympathetic system (LCINEC Symp. found in patients with FMS.
Syst.) (right). CRH = corticotropin-releasing hormone; AVP = argi- Stress system plasticity: neural adaptations to
nine vasopressin; ACTH = adrenocorticotropic hormone; GABA = neonatal pain and stress. Genetic and environmental
y-aminobutyric acid; BZD = benzodiazepines; POMC = pro-
factors interact throughout development to shape the
opiomelanocortin; SP = substance P. (From ref. 28.)
phenotype of an individual. Adverse early experiences,
including abuse, neglect, or severe childhood illness, and
associated medical treatment may influence vulnerabil-
the adrenal medulla secretes epinephrine and norepi- ity to a variety of physio- and psychopathologies, and
nephrine. possibly to disorders such as FMS. Changes in adult
T h e H P A axis and the locus ceruleusi behavior, HPA axis function, central gene expression,
norepinephrine sympathetic nervous system innervate pain threshold, and drug-seeking behavior have been
and interact with each other and other systems in the characterized in male Long Evans rats exposed to either
brain (Figure 3). Thus, CRH neuron projections activate repeated pain (forelimb needle puncture) or maternal
catecholaminergic sympathetic nuclei and vice versa, separation (180 minutes daily) as neonates in the first 2
resulting in a positive reverberating loop. Also, neuro- weeks of life (34). Neonatal exposure to either maternal
transmitters and neuropeptides inhibit the neurons that separation or pain resulted in increased pre- and post-
produce them, via ultrashort feedback loops; tricyclic pubertal pain thresholds in a standard hot-plate test and
antidepressive and central antihypertensive agents exert reduced resistance to infection. Neonatal rats exposed to
their effects at this level. Some neurotransmitters, such maternal separation and pain demonstrated enhanced
as serotonin and acetylcholine, activate both the HPA central CRF neurocircuit activity, HPA axis responsive-
axis and the sympathetic system, whereas y- ness, anxiety-like behavior, and reactivity of the mesen-
aminobutyric acid, the benzodiazepines, and several of cephalic dopaminergic system. A detrimental cascade
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1933

may be initiated by adverse early experience that occurs rine in the sympathetic nervous system, are low in
within a critical period corresponding to a particular patients with FMS (33,39). Another neuroendocrine
dcvelopmental level of the CNS. This cascade may be system, the growth hormone axis, is abnormal in FMS
initiated by the morphogenic actions of serotonin and patients (40,41). However, our understanding of how the
other neurotransmitters in the developing CNS in re- growth hormone and HPA axes are interrelated in FMS
sponse to incoming stimuli. is incomplete.
Hypothalamic-pituitary-gonadal (HPG) axis: Clinical features of FMS, such as widespread pain
stress-induced gonadal compromise. Interactions be- and fatigue, could be related to these observed neuroen-
tween the HPA and HPG axes in FMS patients have yet docrine and sympathetic nervous system perturbations.
to be thoroughly investigated. However, stress has been This hypothesis is supported by the observation that
shown to compromise fertility (35,36). In women, stress- interventions found to be useful in FMS patients affect
related increases in cortisol secretion suppress hypotha- the function of stress-response systems. Further clarifi-
lamic gonadotropin-releasing hormone (GnRH) input cation of neuroendocrine axis function and the relation-
to the pituitary-ovarian axis. Profound and persistent ship of these systems to clinical symptoms may lead to
loss of GnRH input manifests as amenorrhea, whereas improved therapeutic strategies for FMS.
episodic or milder interruptions in GnRH secretion can
bc clinically occult and may present only as decrements
Sleep segment
in luteal-phase progesterone secretion in the setting of
preserved menstrual cycle intervals. Stress management Sleep control mechanisms. Falling asleep is char-
and reduction (such as CBT techniques) may possibly acterized by an advance through stages I, 11,111, and IV
restore GnRH drive, as well as ovulation, menses, and of non-rapid eye movement (non-REM) sleep. As sleep
fertility; this is currently under investigation (Berga S: progresses, there is a series of brief REM periods; sleep
personal communication). becomes more shallow and REM sleep predominates.
Sympathetic nervous and adrenomedullary hor- During waking, alpha waves appear as rapid oscillations
monal systems: differential responses to stressors. The on electroencephalogram (EEG). As sleep is initiated,
major stress response systems, the HPA axis and the the amplitude becomes much smaller, and there is a
sympathetic nervous system, do not necessarily respond mixed-frequency oscillation. In contrast, during the
in the same way to particular stressors (37). In addition, deepest stage of sleep (stage IV), large slow (delta)
the 2 major sympathetic nervous system pathways can be waves occur.
regulated differentially. Interactions between the HPA i n recent years, a rapid increase in knowledge of
axis and the components of the sympathetic nervous basic mechanisms has revolutionized our understanding
system in FMS have not been delineated. of sleep (42). Initially, REM sleep was more readily and
Neuroendocrine findings in patients with FMS. extensively studied. Transection experiments in animals
Patients with FMS exhibit disturbances of the major demonstrated that the basic rhythmic generator for
stress-response systems, the HPA axis and the sympa- REM sleep is in the mammalian brain stem (Figure 4).
thetic nervous system. integrated basal (24-hour urine) In FMS, slow-wave sleep abnormalities appear to
free cortisol levels are low in FMS patients, while plasma be more important than REM sleep. Sleep researchers
cortisol levels obtained at discrete times of the day are are interested in the role of the hypothalamus in slow-
normal (peak) or elevated (trough) (33). Research on wave sleep, how REM sleep and slow-wave sleep are
alterations in basal HPA axis activity is directed toward linked, and the mechanisms of slow-wave sleep produc-
observing the pulsatile characteristics of ACTH and tion. Hypothalamic lesions may cause insomnia, tumors
cortisol secretion. A unique pattern of stimulated HPA may cause hypersomnia, and cells recorded sporadically
axis activity has been noted in patients with FMS. It is throughout the hypothalamus and basal forebrain are
characterized by exaggerated ACTH response to exog- selectively active during slow-wave sleep. However, the
enous CRH, or to endogenous activators of CRH re- specific cells responsible have not been identified.
lease such as insulin-induced hypoglycemia (33,38). The Because FMS may be associated with frequent or
cortisol response to increased ACTH in these stress early awakening, an increased understanding of the
paradigms, and to exercise, is blunted (33,38). mechanisms involved in awakening may have implica-
Patients with FMS may also have disturbed sym- tions with regard to FMS. In animal studies, ventrolat-
pathetic system activity. For example, levels of plasma era1 preoptic (VLPO) neurons may be important in the
neuropeptide Y, a peptide co-localized with norepineph- control of forebrain waking mechanisms, especially in
1934 PILLEMER ET AL

when transferred to normal animals. Several sleep-


Plane of Pontine Forebrain regulatory humoral agents have been identified, includ-
Transection Cortex
’\ ing interleukin-1 (IL-1) and tumor necrosis factor a
(TNFa). Administration of IL-1 or TNFa increases
intensity and duration of non-REM sleep in mice. In
contrast, IL-1 or TNFa blockade inhibits spontaneous
non-REM sleep in rabbits. Blockade of IL-1 or TNFa
prior to experimental manipulations of sleep depriva-
tion, acute mild increases in ambient temperatures, or
injection of microbial products abolishes the expected
sleep responses. TNFa and IL-lP and their receptors
are normal brain products. Further, their messenger
RNA (mRNA) levels or protein levels vary with the
/ Brain Stem
sleep-wake cycle (e.g., rat hypothalamic TNFa and
Figure 4. Rapid eye movement (REM) slecp regulation: representa- TNFa mRNA are highest just after light onset) (45).
tion of a mammalian brain, demonstrating nuclei involved in REM Knockout strains of mice lacking either the 1L-1 type I
sleep. Oblique line represents the plane of transection that preserves
receptor or the 55-kd TNFa receptor sleep less than
the signs of REM caudally. The REM-promoting neurons primarily
contain the neurotransmitter acetylcholine. Two nuclei found at the their strain controls. In addition, changes in sleep regu-
junction of the midbrain and pons, the laterodorsal tegmental (LDT) lation that may be beneficial to the host occur during
and pendunculopontine tegmental (YPT) nuclei, project to target areas infection. These sleep responses are induced by specific
in the reticular formation that control the rapid eye movements of microbial components, e.g., bacterial cell wall muramyl
REM sleep, and the muscle atonia zones that control the muscle
peptides and lipopolysaccharide or influenza vital
atonia of REM sleep. Cholinergic agonists placed in this region
produce a state that simulates REM sleep. Firing of serotonergic double-stranded RNA. These microbial products also
dorsal raphc nucleus (DRN) neurons and norepinephrinergic locus induce cytokine production, and their somnogenic ac-
ceruleus (LC) neurons slows during slow-wave sleep and virtually tions are attenuated if IL-1 or TNFa is inhibited. The
ceases just before and during REM sleep. This suggested that these findings cited above strongly implicate the immune
neurons might disinhibit the cholinergic LDTiPPT neurons, a theory
that was later tested and shown to be correct. At sleep onset,
response modifiers IL-1 and TNFa in sleep regulation.
cholinergic, REM-on neurons are initially quiescent, then become The humoral regulation of sleep involves com-
active, and at a certain threshold, REM sleep occurs. The cycle repeats plex cascades of biochemical events within the brain,
throughout the night, and the increasing duration of REM periods analogous to events involved in immune cell regulation.
reflects circadian modulation. REM-suppressive neurons, containing For example, IL-1 induces TNFa and growth hormone-
norepinephrine and serotonin, have an opposite discharge pattern, and
as they turn off, the cholinergic neurons bccome active. BRF, PRF,
releasing hormone (GHRH) production, while anti-
and MRF = bulbar, pontine, and mesencephalic reticular formation. GHRH inhibits IL-1-induced sleep (46). Both IL-1 and
(Adapted, with permission, from Serninurs in the Neurosciences [42].) TNFa induce nitric oxide (NO) production. In contrast,
inhibition of NO synthase inhibits spontaneous sleep
and IL-1-induced sleep. Administration of exogenous
histaminergic tuberomamillary nucleus cells (43). The NO donors, such as S-nitroso-N-acetylpenicillamine or
Fos protein, which accumulates in recently activated molsidomine, enhances sleep. Data such as these, cou-
neurons, has been used in immunocytochemical studies pled with the recent availability of knockout strains of
to show that sleep-activated VLPO neurons innervate mice lacking one or more of the genes included in the
the tuberomammillary nucleus. These studies suggested IL-1 and TNFa families of molecules, will facilitate
that sleep-wake states may be regulated by this mono- research into the biochemical events and pathways in-
synaptic pathway in the hypothalamus. Ongoing struc- volved in sleep regulation.
tural studies suggest the exciting possibility that these Sleep deprivation influences on behavior and
neurons may coordinate other functions including brain immune function in animals. The effects of profound
stem nuclei, such as those involved in pain perception, sleep deprivation resemble the effects of loss of a vital,
and perhaps endocrine projections, such as those in- physical requirement, such as food (47). In young
volved in the stress system. healthy humans, short-term sleep deprivation results in
Animal models of sleep. Sleep is regulated in part 1) profound decrements in cognitive function, 2) pro-
by humoral agents (44). Sleep-promoting substances gressive decreases in oral temperature, 3) behavioral-
accumulate in CSF during wakefulness and induce sleep psychiatric changes (irritability, delusions), 4) nonspe-
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1935

cific neurologic signs, and 5) increased sensitivity to pain old rats could not be accounted for by sex steroid
(48). The specific organic changes due to sleep depriva- hormones in females. Similar studies in male rats also
tion or disruption in individuals with compromised excluded the possibility of a hormonal role. Further
health have not been elucidated, but may exacerbate investigation demonstrated that the instability of ther-
disease and hinder recovery. mal and sleep rhythms with aging occurs independently,
Prolonged sleep deprivation in rats produces i.e., the unstable thermal and sleep regulatory rhythms
syndromic physical changes that occur in 2 stages over are not linked to one another (55). Thus, although sleep
time, and culminate in death. The first stage is longer is linked to temperature regulation in younger animals,
and is marked by a progressive negative energy balance, this may change under certain conditions, including
accompanied by a mild elevation in body temperature aging.
and its subsequent return to baseline. The second phase Roles of sleep and circadian rhythm in neuro-
is heralded by a breakdown in host defense, manifested endocrine function. Circadian rhythmicity is an intrinsic
as systemic infection by lethal opportunistic organisms endogenous signal that, in mammals, is generated by the
and a mild drop in body temperature. This second stage hypothalamic suprachiasmatic nucleus (56). Although
is acute and is characterized by a cachectic-like state of the circadian rhythm is an endogenous signal, it is
dwindling health, suggestive of cytokine- and endotoxin- influenced by environmental periodicities, such as light-
mediated septicemia and shock. dark cycle (56,57). The sleep-wake cycle can also exert
Detection of lethal opportunistic organisms in synchronizing effects on the suprachiasmatic nucleus,
the blood of sleep-deprived rats is diagnostic of a which integrates environmental and sleep-wake infor-
functional impairment that implicates the immune sys- mation and drives a variety of endocrine, behavioral,
tem (49). Despite systemic infection, sleep-deprived rats and metabolic rhythms. Measurement of hormones at a
do not exhibit fever or marked tissue inflammatory single time of day has little meaning, since the endocrine
reactions typical of most infectious disease states, indi- system displays intricate temporal organization and re-
cating that sleep deprivation is immunosuppressive. alizes different states of endocrine function at different
Whether such immunosuppression is a primary or a times of day (58).
secondary effect of sleep deprivation is unknown, al- In FMS, slow-wave sleep may be interrupted by
though several signs of sleep deprivation are consistent intrusions of alpha activity (59). These intrusions may be
with early changes in immunocompetency. Furthermore, important, because most of the endocrine and cardio-
responses by immunomodulators to host defense chal- vascular effects of sleep are observed during deep sleep,
lenges during sleep deprivation may mediate part of the including secretion of growth hormone and prolactin
deep negative energy balance that leads to a cachectic- and inhibition of corticotropic activity and thyroid-
like state. stimulating hormone secretion.
Thermal regulation and sleep-wake physiology. Understanding sleep control mechanisms may
Thermal stress has an extremely important influence on ultimately lead to a better understanding of the sleep
sleep (50,51). RBM sleep is sensitive to ambient tem- disorder associated with FMS. The timing, duration, and
perature. Basal forebrain lesions in rats cause insomnia quality of sleep are controlled by 2 major components,
and interfere with thermoregulation, although at higher the homeostatic and circadian components (60). The
ambient temperatures REM sleep may appear normal. homeostatic component reflects the amount of sleep
Pharmacologic interventions may also interfere with required to compensate for the duration of wakefulness
sleep; phentolamine, an a1 -noradrenergic antagonist, and may primarily control slow-wave sleep. In contrast,
decreases sleep in rats in a similar way to that seen in the the circadian component is thought to primarily regulate
presence of lesions in the basal forebrain, when ambient REM sleep, as well as to exert a modulatory effect on
temperature is not taken into account (52). the timing and duration of sleep. Regardless of the
Experiments in female rats have demonstrated presence or timing of sleep, the cortisol pattern shows
that aging is also associated with thermal and sleep little modification (circadian dependent). In contrast,
rhythm disturbances (53). However, when old anestrous growth hormone disappears when there is no sleep, and
female rats were compared with young oophorecto- reappears with the replacement of sleep (sleep depen-
mized rats, the oophorectomized animals did not show a dent). The circadian system may be involved in the
deterioration of rhythm (54). In contrast, they showed waking function and altering the expression of the
more precisely regulated rhythms, suggesting that the homeostatic component; intrusion of physiologic waking
instability of thermal and sleep rhythms developing in during sleep in FMS could be related to an altered
1936 PILLEMER ET AL

relationship between the homeostatic and circadian unrefreshing sleep, but they report more depressive
components. symptoms and psychological distre
Sleep, particularly slow-wave sleep, has a role in studies show that the alpha EEG sleep anomaly and
controlling the release of growth hormone (61). In symptom5 may follow a febrile episode.
normal young subjects, the amount of growth hormone The theoretical model that FMS patients have
secretion demonstrates a dose-response relationship to disordered chronobiologic functions that are linked to a
the amount of slow-wave sleep. In contrast, awakening dysregulation of their sleep-waking brain is based on the
during sleep while growth hormone is being secreted following evidence (65-67): 1) research finding\ on the
results in a rapid inhibition o f growth hormone secre- disordered sleep-wake physiology, symptoms, and be-
tion. In FMS, sleep fragmentation that manifests as havior in patients with FMS and chronic fatigue syn-
awakening, arousal, or perhaps alpha intrusion would be drome; 2) cxperimcntal studies that link the neurochem-
expected to inhibit growth hormone secretion (62). ical, eytokine-immune-neurocndoerine thermal systems
Studies in patients with FMS have shown a decrease in and the sleep-wake cycle in animals and humans; 3)
insulin-like growth factor 1, the hormone secreted by the studies and preliminary data on the interrelationships of
liver, which could possibly occur in response to de- sleep-wakefulness, IL-1 , and aspects of the peripheral
creased growth hormone stirnulation (40.63). immune and neuroendocrine functions in healthy men
Aging has differential effects on sleep and endo- (in women t h e w sleep-wake-related immune-
crine system components (55,56). In terms of slow-wave neuroendocrine functions differ between high and low
sleep decline and growth hormone secretion, we are old progesterone phases of the menstrual cycle); and 4) the
by age 40; whereas REM sleep and the progressive rise observations of alterations in aspects of neurochemical
of evening cortisol levels change at a slower ratc over a (serotonin, SP), immune, neuroendocrine, and auto-
lifetimc (64). Restoration of deep (slow-wave) sleep and nomic functions in patients with FMS.
its endocrine correlates has been studied in healthy Time series analyses o f measures of the circadian
volunteers. The majority of currently available hypnotic patterns of the sleep-wakefulness, neurochemical, and
agents do not increase slow-wave sleep. It may be worth cytokine-immune-neuroendocrine-thermal functions in
investigating whether alpha suppression in FMS can be patients with FMS and chronic fatigue syndrome should
overcome by stimulation of slow-wave \leep, and determine whether alterations in aspects of these bio-
whether such stimulation has a beneficial effect. logic operatiom accompany the disordered sleep-wake
FMS as a model for sleep dysregulation. Disor- physiology that results in nonrefreshing sleep, chronic
dered sleep physiology is accompanied by nonrestorative pain, fatigue, and cognitive and behavioral symptoms.
sleep, fatigue, diffuse musculoskeletal pain, and There is evidence, based on diary reports, of a bidirec-
cognitive-behavioral symptoms in patients with FMS tional within-person association between sleep quality
(59). Clinical studies show that measures of pain and and pain in women with FMS (68), i.e., nights of
fatigue are related to ratings of poor sleep. Also, non- relatively poor 4eep are followed by significantly greater
rcfreshing sleep in patients with FMS is commonly pain the next day, and more painful days are followed by
associated with altered sleep physiology. These physio- poorer sleep at night. However. the biologic changes
logic disturbances include the alpha (7-12-IIz) EEG underlying this relationship have not been determined.
sleep anomaly and sleep-related features of periodic
involuntary limb movements, periodic K-alpha EEG,
Conclusion
and apnea-hypopnea in subgroups of patients. Experi-
mental disruption of slow-wave sleep with noise artifi- Fibromyalgia has been regarded by many as a
cially induces the alpha EEG sleep anomaly, nonrefresh- diffuse and nebulous syndrome that is “all in the pa-
ing sleep, diffuse myalgia, and fatigue in healthy tient’s head.” There is mounting evidence that these
subjects. Patients with FMS have diurnal impairment in impressions have an element of truth in them-not in a
cognitive performance functions that arc sensitive to way that disparages FMS patients, but at the physiologic
sleep deprivation. They show a reduction in the speed, level. Much of the symptomatology of FMS is familiar in
but not the accuracy, of complex performance tasks other contexts to researchers in the areas of pain,
carried out hourly throughout the day, and ratc them- neuroscience, endocrinology, and sleep, particularly
selves as sleepy and fatigued. In comparison with FMS where centrally mediated mechanisms are concerned.
patients, patients with chronic somatoform pain disorder During the workshop, many of the intricate in-
do not show the alpha EEG sleep anomaly or describe teractions between components of the central nervous
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1937

and endocrine systems were discussed in relation to neuromechanisms of pain, neuroendocrine functioning,
pain, circadian rhythms, and sleep. These systems are and immune alterations that might relate to FMS.
responsive to stressors and are functionally, anatomi- The workshop generated considerable excite-
cally, and physiologically intertwined. Furthermore, ment among the participants from diverse fields, who
there is clear evidence of sex differences in the developed some mutual understanding regarding the
regulation of pain and sleep, as well as in circadian interrelationships among pain, neuroendocrine func-
rhythms. Also, circadian rhythms and their relation- tions, circadian rhythms, and sleep. We believe that an
ship to homeostatic mechanisms of sleep appear to interdisciplinary approach will accelerate progress in
alter with increased age. research into FMS and other related disorders.
In the field of pain related to FMS, we must
devote more effort to the development of animal models
ACKNOWLEDGMENTS
and to the understanding of CNS functions. In addition,
the psychosocial factors involved in pain vulnerability The authors and cosponsors thank the following pre-
and pain expression require clarification. Better under- senters for their materials and efforts that contributed to this
standing of pain transmission and pain inhibition may Workshop Summary: Dr. Sarah L. Berga, Dr. Daniel J. Clauw,
Dr. Ronald Dubner, Dr. Richard H. Gracely, Dr. Carol A.
lead to new treatments in FMS. Everson, Dr. David S. Goldstein, Dr. Francis J. Keefe, Dr.
Questions regarding neuroendocrine function in Robert W. McCarley, Dr. Paul M. Plotsky, Dr. 1. Jon Russell,
FMS remain unanswered. How do we distinguish vul- Dr. Evelyn Satinoff, Dr. Wendy F. Sternberg, and Dr. Eve Van
nerability in neuroendocrine function leading to the Cauter. We also thank Dr. Kenneth Casey, Dr. James M.
development of FMS versus abnormalities that might Krueger, and Dr. Susana A. Serrate-Sztein, who served with
result from FMS? How are gonadal hormones and aging the authors on the Planning Committee. We are grateful for
the assistance of the Conference Logistics Team of the Na-
related to FMS? Findings from neuroendocrine and tional lnstitute of Arthritis and Musculoskeletal and Skin
pain research must be integrated in order to understand Diseases.
how they interact in FMS. Neuroendocrine function
studies should take into account the importance of the
complex temporal variations in hormone levels. REFERENCES
In the field of sleep research, we are beginning to Wolfe F, Smythe HA, Yunus MB, Bennett RM, Bombardier C,
address the fundamental mechanisms involved in sleep- Goldenberg DL, Tugwell P, Campbell SM, Abeles M, Clark P,
Fam AG, Farber SJ, Fiechtner JJ, Franklin CM, Gatter RA,
wake physiology, the rhythms of the body, and the Hamaty D, Lessard J, Lichtbroun AS, Masi AT, McCain GA,
effects of the dysregulation of these rhythms on FMS. Reynolds WJ, Romano TJ, Russell IJ, Sheon RP: The American
Researchers are shifting attention from the brain stem College of Rheumatology 1990 criteria for the classification of
fibromyalgia: report of the multicenter criteria committee. Arthri-
to the hypothalamus as an area of importance in tis Rheum 33:160-172, 1990
regulation. Willis WD, Coggeshall RE: Sensory Mechanisms of the Spinal
The rhythms of the body and sleep-wake physi- Cord. Second edition. New York, Plenum Press, 1991
Chen ACN: Human brain measures of clinical pain: a review. I.
ology are intimately connected to cytokines and aspects Topographic mappings. Pain 54:115-132, 1993
of neuroendocrine functioning. Further investigation Chen ACN: Human brain measures of clinical pain: a review. 11.
into the ways in which sleep deprivation is involved in Tomographic imagings. Pain 54:133-144, 1993
Dubner R, Basbaum A: Spinal dorsal horn plasticity following
problems of repair or lack of repair may be useful in tissue damage and nerve injury. In, Textbook of Pain. Edited by R
understanding FMS. Additional research is needed to Melzack, PD Wall. New York, Churchill-Livingstone, 1994
clarify the role of thermal factors and their intimate Dubner R, Ruda MA: Activity-dependent neuronal plasticity
following tissue injury and inflammation. Trends Neurosci 15:9h-
linkage to sleep-wakefulness, the rhythms of the body, 103, 1992
and aging in the development of sleep abnormalities in Bennett GJ: Neuropathic pain. In, Textbook of Pain. Edited by R
FMS. Future investigation may involve differences in Melzack. PD Wall. New York. Churchill-Livingstone, 1949
8. Aaron LA, Bradley LA, Alarcon GS, Alexander RW, Triana-
males and females, and estrus versus nonestrus systems Alexander M, Martin MY, Alberts KR: Psychiatric diagnoses in
involved in animal models. Little information is avail- patients with fibromyalgia are related to health care-seeking
able, not only with regard to normal physiology of behavior rather than to illness. Arthritis Rheum 39:436-445, 1996
9. Bradley LA, Alarcon GS, Alexander RW, Alexander MT, Aaron
thermal mechanisms, but also with regard to how these LA, Alberts KR, Martin MY: Abnormal central processing of
mechanisms might be impaired in FMS. Finally, there is dolorimeter stimuli in patients and community residents with
much work to be done in the area of sleep physiology fibromyalgia: one year reliability (abstract). Arthritis Rheum 38
(suppl 9):S318, 1995
and its alterations; the findings of such investigations 10. Russell IJ, Orr MD, Littman B, Vipraio GA, Alboukerk D,
may allow associations with the results of studies on Michalek JE, Lopez Y, MacKillip F: Elevated cerebrospinal fluid
1938 PILLEMER ET AL

levels of substance P in patients with the fibromyalgia syndrome. 30. Vanderpool J, Rosenthal N, Chrousos GP, Wchr T, Skcwerer R,
Arthritis Rheum 37:1593-1601, 1994 Kasper S, Gold PW: Evidence for hypothalamic CRH deficiency in
11. Bradley LA, Alberts KR, Alarcdn GS, Alexander MT, Mountz JM, patients with seasonal affective disorder. J Clin Endocrinol Metab
Weigent DA, Liu HG, Blalock JE, Aaron LA, Alexander RW, San 7211382-1387, 1991
Pedro EC, Martin MY, Morel1 AC: Abnormal brain regional 31. Demitrack M, Dale J, Straus S, Laue L, Listwak S, Kruesi M,
cerebral blood flow and cerebrospinal fluid levels of substance P in Chrousos GP, Gold PW: Evidencc of impaired activation of the
patients and non-patients with fibromyalgia (abstract). Arthritis hypothalamic-pituitary-adrenal axis in patients with chronic fa-
Rheum 39 (suppl 9):S212, 1996 tigue syndrome. J Clin Endocrinol Metab 73:1224-1234, 1991
12. Russell IJ: Neurochemical pathogenesis of fibromyalgia syndrome. 32. Magiakou MA, Mastorakos G, Rabin D, Dubbert B, Gold PW,
J Musculoskeletal Pain 4:61-92, 1996 Chrousos GP: Hypothalamic corticotropin relcasing hormone
13. Mountz JM, Bradley LA, Model1 JG, Alexander RW, Triana- suppression during the postpartum period: implications for the
Alexander M, Aaron LA, Stewart KE, Alarcdn GS, Mountz JD: increase of psychiatric manifestations during this timc. J Clin
Fibromyalgia in women: abnormalities of regional cerebral blood Endocrinol Metab 81:1912-1917, 1996
flow in the thalamus and the caudate nucleus are associated with 33. Crofford LJ, Pillemer SR, Kalogcras KT. Cash JM, Michelson D,
low pain threshold levels. Arthritis Rheum 38:926-938, 1995 Kling MA, Sternberg EM, Gold PW, Chrousos GP, Wilder R L
14. Iadorola MJ, Max MB, Berman KF, Byas-Smith MG, Coghill RC, Hypothalamic-pituitary-adrenal axis perturbation in patients with
Gracely RH, Bennett GJ: Unilateral decrease in thalamic activity fibromyalgia. Arthritis Rheum 37:1583-1592, 1994
observed with positron emission tomography in patients with 34. Plotsky PM, Meaney MJ: Early, postnatal expcricncc altcrs hypo-
chronic neuropathic pain. Pain 6355-64, 1995 thalamic corticotropin-releasing factor (CRF) mRNA, median
15. DiPiero V, Jones AKP, Ianotti F: Chronic pain: a PET study of the eminence CRF content and stress-induced release in adult rats.
central effects of percutaneous high cervical cordotomy. Pain Brain Res Mol Brain Res 18:195-200, 1993
46:9-12, 1991 35. Berga SL, Mortola JF, Girton L, Suh B, Laughlin G, Pharn P, Yen
16. Hsieh J, Belfrage M, Stone-Elander S, Hansson P, Ingvar M: SS: Neuroendocrine aberrations in womcn with functional hypo-
Central representation of ongoing neuropathic pain studies by thalamic amenorrhea. J Clin Endocrinol Metab 68:30 1-308, 1989
positron emission tomography. Pain 63:225-236, 1995 36. G i l a DE, Berga SL: Cognitive and psychiatric correlates of
17. Feine JS, Bushnell MC, Miron D, Duncan GH: Sex differences in functional hypothalamic amenorrhea: a controlled comparison.
the perception of noxious heat stimuli. Pain 44:255-262, 1991 Fertil Steril 60:486-492, 1993
18. Notermans SL, Tophoff MM: Sex differences in pain tolerance 37. Pacak K, Palkovits M, Kopin IJ, Goldstein DS: Stress-induced
and pain apperception. Psychiatr Neurol Neurochir 70:23-29. 1967 norepinephrine release in the hypothalamic paraventricular nu-
19. Romero MT, Bodnar RJ: Gender differences in two forms of cleus and pituitary-adrenocortical and sympathoadrenal activity:
cold-water swim analgesia. Physiol Behav 372393-897, I986 in vivo microdialysis studies. Front Neuroendocrinol 1689-1 50,
20. Kavaliers M, Innes D: Stress-induced analgesia and activity in deer 1995
mice: sex and population differences. Brain Res 425:49-56, 1987 38. Griep EN, Boersma JW, de Kloet EP: Altcred reactivity of the
21. Kepler KL, Kest B, Kiefel JM, Cooper ML, Bodnar RJ: Roles of hypothalamic-pituitary-adrenal axis in the primary fibromyalgia
gender, gonadectomy and estrous phase in the analgesic effects of syndrome. J Rheumatol 20:469-474, 1993
intracerebroventricular morphine in rats. Pharmacol Biochem 39. Clauw DJ, Sabol M, Radulovic D, Wilson B, Katz P, Baraniuk J:
Behav 34:119-127, 19x9 Serum neuropeptides in patients with both FMS and chronic
22. Mogil JS, Sternberg WF, Kest B, Marek P, Liebeskind JC: Sex fatigue syndrome (abstract). J Musculoskeletal Pain 3 (suppi 1):79,
differences in the antagonism of swim stress-induced analgesia: 1995
effects of gonadectom and estrogen replacement. Pain 53:17-2.5, 40. Bennett RM, Clark SR, Campbell SM, Burkhardt CS: Low levels
1993 of somatomedin C in paticnts with the fibromyalgia syndrome: a
23. Lorig KR, Mazonson PD, Holman HR: Evidence suggesting that possible link between sleep and muscle pain. Arthritis Rheum
health education for self-management in patients with chronic 35:1113-1116, 1992
arthritis has sustained health benefits while reducing health care 41. Griep EN, Boersma JW, de Kloet E:R: Pituitary release of growth
costs. Arthritis Rheum 36:439-446, 1993 hormone and prolactin in the primary fibromyalgia syndrome.
24. Parker JC, Smarr KL, Buckelew SP, Stucky-Ropp RC, Hewett JE, J Rheumatol 21:2125-2130, 1994
Johnson JC, Wright GE, Irvin WS, Walker SE: Effects of stress 42. McCarley RW, Greene RW, Rainnie D, Portas CM: Brainstem
management on clinical outcomes in rheumatoid arthritis. Arthri- neuromodulation and REM sleep. Semin Neurosci 7:341-354,
tis Rheum 38:1807-1818, 1995 199.5
25. Keefe FJ, Caldwell DS, Williams DA, Gill KM, Mitchell D, 43. Sherin JE, Shiromani PJ, McCarley RW, Saper CH: Activation of
Robertson C, Martinez S, Nunley J, Beckham JC, Crisson JE, ventrolateral preoptic neurons during sleep. Science 271 :216-219,
Helms M: Pain coping skills training in thc management of 1996
osteoarthritis knee pain: a comparative study. Behav Ther 21 :49- 44. Krueger M, Majde A Microbial products and cytokines in sleep
62, 1990 and fever regulation. Crit Rev Immunol 14:355-379, 1994
26. White KP, Nielson WR: Cognitive behavioral treatment of fibro- 45. Fang J, Kapis J, Seyer JM, Wang Y, Krueger JM: The TNF 55kD
myalgia syndrome: a followup assessment. J Rheumatol 22:717- receptor and the IL-1 type I receptor are involved in physiological
721, 1995 sleep regulation (abstract). Soc Neurosci Abstracts 22: 147, 1996
27. Vlaeyen JW, Teeken-Gruben NJ, Goossens ME, Rutten-van 46. Obi1 F Jr, Fang L, Paync LC, Krueger JM: Growth hormone-
Molkcn MP, Pelt RA, van Eek H, Heuts PH: Cognitive-education releasing hormone (GHRH) mediates the sleep-promoting activ-
treatment of fibromyalgi randomized clinical trial. I. Clinical ity in interleukin-1 (IL-1) in rats. Neurocndocrinology 61:559-565,
cffects. J Rheumatol 23:1237-1245, 1906 1995
28. Chrousos GP, Gold PW: The concepts of stress and stress system 47. Everson C A Functional consequences of sustained sleep dcpriva-
disorders: overview of physical and behavioral homeostasis. JAMA tion in the rat. Behav Brain Res 69:43-54, 1995
267:1244-1252, 1992 48. Everson C A Clinical manifestations of sleep deprivation. In, Sleep
29. Chrousos GP: The hypothalamic-pituitary-adrenal axis and Science: Integrating Basic Research and Clinical Practice (Mono-
immune-mediated inflammation. N Engl J Med 332:1351-3362, graphs in Clinical Neuroscience). Edited by WJ Schwartz. Basel,
1995 Karger (in press)
FMS NEUROSCIENCE AND ENDOCRINOLOGY WORKSHOP 1939

49. Everson C A Sustained sleep deprivation impairs host defense. regulation in young poor sleepers. Neurophysiol Clin 26:40-50,
Am J Physiol 265:R114X-R1154, 1993 1996
50. Szymusiak R, Satinoff E: Maximal REM sleep time defines a 61. Van Cauter E, Plat L Physiology of growth hormone secretion
narrowcr thermoneutral zone than does minimal metabolic rate. during sleep. J Pediatr 128:S32-S37, 1996
Physiol Behav 26587-690, 1981 62. Jacobsen S, Main K, Danneskiold-Samsoe B, Skakkebaek NE: A
51. Satinoff E: Thermal influences on REM slecp. In, Clinical Physi- controlled study on serum insulin-like growth factor-I and urinary
ology of Sleep. Edited by R Lydic, J Biebuyck. New York, Oxford excretion of growth in fibromyalgia. J Rheumatol 22:1138-1140,
University Press, 1988 1995
52. Kent S, Satinoff E: Influence of ambient temperature on sleep and
63. Copinschi G, van Cauter E: Effects of ageing on modulation of
body temperature after phentolamine in rats. Brain Res 511:227-
233, 1990 hormonal secretions by sleep and circadian rhythmicity. Horm Res
53. Li H, Satinoff E: Changes in circadian rhythms of body tempera- 4320-24, 1995
ture and sleep in old rats. Am J Physiol 269:R208-R214, 1995 64. Van Cauter E, Leproult R, Kupfer DJ: Effects of gender and age
54. Li H, Satinoff E: Body temperature and sleep in intact and on the levels and circadian rhythmicity of plasma cortisol. J Clin
ovariectomized female rats. Am J Physiol 40:R1750-R1758, 1996 Endocrinol Metab 81:2468-2473, 1996
55. Satinoff E, Li H, Tcheng TK, Liu C, McArthur AJ,Medanic M, 65. Moldofsky H: Sleep, neuroimmune and neuroendocrine functions
Gillette MU: Do the suprachiasmatic nuclei oscillate in old rats as in FMS and chronic fatigue syndrome. Adv Neuroimmunol 5:39-
they do in young ones? Am J Physiol 265:R1216-R3222, 1993 56, 1995
56. Murphy PJ, Campbell SS: Physiology of the circadian system in 66. Moldofsky H: Chronobiological influences on fibromyalgia syn-
animals and humans. J Clin Neurophysiol 13:2-16, 1996 drome: theoretical and therapeutic implications. Baillieres Clin
57. Czeisler CA: The effect of light on the human circadian pace- Rheumatol 8:801-810, 1994
maker. Ciba Found Symp 183954-290, 1995 67. Moldofsky H: Central nervous system and peripheral immune
58. Turek FW: Circadian rhythms. Recent Prog Horm Res 49:43-90, functions and the sleep-wake systcm. J Psychiatr Neurosci 19:368-
1994 374, 1994
59. Moldofsky H: Sleep and fibrositis. Rheum Dis Clin North Am 68. Affleck G, Urrows S, Tennen H, Higgins P, Abeles M: Sequential
15:91-103, 1989 daily relations of sleep, pain intensity, and attention to pain among
60. Benoit 0, Aguirre A: Homeostatic and circadian aspects of sleep women with fibromyalgia. Pain 68:363-368, 1996

You might also like