You are on page 1of 26

HPA Axis-Rhythms

Francesca Spiga,1 Jamie J. Walker,1,2 John R. Terry,2 and Stafford L. Lightman*1

ABSTRACT
The hypothalamic-pituitary-adrenal (HPA) axis regulates circulating levels of glucocorticoid hor-
mones, and is the major neuroendocrine system in mammals that provides a rapid response and
defense against stress. Under basal (i.e., unstressed) conditions, glucocorticoids are released
with a pronounced circadian rhythm, characterized by peak levels of glucocorticoids during the
active phase, that is daytime in humans and nighttime in nocturnal animals such as mice and
rats. When studied in more detail, it becomes clear that the circadian rhythm of the HPA axis is
characterized by a pulsatile release of glucocorticoids from the adrenal gland that results in rapid
ultradian oscillations of hormone levels both in the blood and within target tissues, including the
brain. In this review, we discuss the regulation of these circadian and ultradian HPA rhythms, how
these rhythms change in health and disease, and how they affect the physiology and behavior of
the organism.  C 2014 American Physiological Society. Compr Physiol 4:1273-1298, 2014.

Introduction circulation, glucocorticoids access target tissues, such as the


liver, the heart and vascular tissues, to exert metabolic and
The HPA axis cardiovascular effects, respectively, and the brain, where for
The hypothalamic-pituitary-adrenal (HPA) axis regulates cir- example, glucocorticoids promote cognitive processes neces-
culating levels of glucocorticoid hormones, and is the major sary to cope with a threatening situation (47).
neuroendocrine system in mammals that provides a rapid Glucocorticoids also regulate the activity of the HPA axis,
response and defense against stress (Fig. 1A). Glucocorti- and thus their own production, through feedback mechanisms,
coids (cortisol in humans and corticosterone in rodents) are acting at the level of the pituitary gland where they inhibit
vital hormones that regulate many physiological functions, ACTH release (91), and at the level of the PVN where they
including glucose, fat, and protein metabolism (32, 121, 138). inhibit the synthesis and release of CRH and AVP (42, 43,
In addition, glucocorticoids exert anti-inflammatory and 91). In addition, glucocorticoids indirectly regulate HPA axis
immunosuppressive actions and can affect mood and cog- activity via modulation of other brain structures, including the
nitive function (35, 49, 128). Under basal (i.e., unstressed) hippocampus, the amygdala, and the prefrontal cortex, which
conditions, the dynamics of the HPA axis is characterized by in turn regulate the activity of the PVN (196).
both a circadian and an ultradian rhythm of hormone secre-
tion (Fig. 1B). In addition to this, the activity of the HPA
axis increases when the organism is exposed to stress. This Adrenal glucocorticoid synthesis
stress response is initiated by the activation of several afferent Glucocorticoids are synthesized in the zona fasciculata of
neural pathways, including those originating from limbic and the adrenal cortex. Glucocorticoid steroidogenesis is acti-
brain-stem structures (196), which in turn induce the release vated when ACTH binds to its specific cell surface G-protein-
of corticotropin-releasing hormone (CRH) and arginine vaso- coupled receptor, the melanocortin type-2 receptor (MC2R)
pressin (AVP) from parvocellular neurons of the hypothala- (137). Upon ACTH binding, MC2R undergoes conforma-
mic paraventricular nucleus (PVN) (62, 159, 167, 197). Upon tional changes that activate adenylyl cyclase, leading to an
stimulation, these neuropeptides are released from neuronal increase in intracellular levels of cyclic adenosine monophos-
terminals at the level of the median eminence into the por- phate (cAMP). This intracellular increase in cAMP activates
tal vessels (6). Via the portal circulation, CRH and AVP downstream signaling pathways, including the protein kinase
reach the anterior pituitary, where they bind to their spe- A (PKA) pathway. Activation of PKA induces acute syn-
cific receptors, CRH receptor 1 (CRHR1) and AVP 1B recep- thesis of glucocorticoids via both genomic and nongenomic
tor (V1bR), respectively (7, 30), to stimulate the release of
* Correspondence to Stafford.Lightman@bristol.ac.uk
adrenocorticotropic hormone (ACTH) from corticotroph cells
1 University of Bristol, Henry Wellcome Laboratories for Integrative
into the general circulation. ACTH, in turn, stimulates the Neuroscience & Endocrinology, Bristol, United Kingdom
adrenal gland to produce and secrete glucocorticoid hor- 2 University of Exeter, College of Engineering, Mathematics and
mones (42, 45). In contrast to peptide hormones, which are Physical Sciences, Exeter, United Kingdom
presynthesized and stored within the cytoplasm and released Published online, July 2014 (comprehensivephysiology.com)
upon activation, glucocorticoids are synthesized de novo upon DOI: 10.1002/cphy.c140003
stimulation of the adrenal. Once released into the blood Copyright  C American Physiological Society.

Volume 4, July 2014 1273


HPA Axis-Rhythms Comprehensive Physiology

Circadian and stress inputs


(A)
(B)
PVN
Hypothalamus
100
CRH and AVP

Corticosterone (ng/mL)
80

Corticotroph cells 60
Pituitary
gland 40
ACTH
20

CORT 0
0900 1500 2100 0300 0900
Clock time (h)
Adrenal gland Adrenal cortex

Figure 1 The hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid rhythms. (A) The hypothalamic
paraventricular nucleus (PVN) receives circadian input from the SCN and stress inputs from the brainstem and
from regions of the limbic system such as the hippocampus and amygdala. The PVN projects to the median
eminence where it releases corticotrophin-releasing hormone (CRH) and arginine vasopressin (AVP) into the
hypothalamic-pituitary portal circulation. CRH passes through this vascular route to access corticotroph cells
in the anterior pituitary, which respond with the rapid release of adrenocorticotropic hormone (ACTH) from
preformed vesicles into the general circulation. In turn, ACTH reaches the adrenal cortex where it activates the
synthesis and secretion of glucocorticoid hormones (CORT). Glucocorticoids regulate the activity of the HPA axis,
and thus their own production, through feedback mechanisms acting at the level of the pituitary gland where they
inhibit ACTH release, and at the level of the PVN where they inhibit the release of CRH and AVP. Reproduced with
permission from (116). (B) Under basal (i.e., unstressed) conditions, the dynamics of glucocorticoid secretion is
characterized by both a circadian (gray line) and an approximately hourly ultradian rhythm (black line). In the
rat, peak levels of corticosterone occur during the active evening phase. Shaded region indicates the dark phase.
Reproduced with permission from (200).

mechanisms. The rate-limiting step in glucocorticoid synthe- stabilized by chaperone molecules (140, 147, 215), including
sis is the mobilization of cholesterol and its transfer inside the the 90-kDa heat-shock protein (HSP-90) (149). Upon glu-
mitochondria, and this process is mediated by the activity of cocorticoid binding, the chaperone-receptor complex dissoci-
the steroidogenic acute regulatory protein (StAR) (118, 185). ates to enable the ligand-receptor complex to translocate to the
Activation of the PKA signaling pathway activates nuclear nucleus, where it functions as transcription factor to modulate
transcription factors, such as the cAMP response element genomic events via activation or repression of transcription
(CRE) binding protein (CREB), which in turn induce tran- of glucocorticoid target genes.
scriptional activation of a number of steroidogenic genes, GR and MR differ in their affinity for glucocorticoids. The
including StAR. In parallel to StAR transcription, PKA also affinity of the endogenous hormones corticosterone and cor-
induces posttranslational modification of StAR, resulting in tisol for MR is approximately 10-fold higher than the affinity
phosphorylation at a specific site that activates StAR and for GR. Therefore, variations in circulating levels of gluco-
increases its ability to transport cholesterol across the mito- corticoids lead to shifts in the balance between MR and GR
chondrial membrane (9). Within the mitochondria, choles- activity (158). The high-affinity MR is activated at very low
terol is subjected to enzymatic modifications by a cascade of concentrations of glucocorticoids, whereas the lower affinity
steroidogenic regulators, which ultimately lead to glucocorti- GR is only activated at high concentrations. In basal (non-
coid synthesis (21, 36, 80). stressed) physiological states, when glucocorticoid concen-
trations are low, MR is activated and localized in the nucleus,
whereas GR remains latent in the cytoplasm. Once gluco-
Glucocorticoid receptors and genomic corticoid concentrations increase above a threshold level, for
responses example during the circadian peak or following exposure to
Glucocorticoids exert their effects by binding to specific stress, GR translocates into the nucleus where it exerts its
receptors expressed in target tissues. Two receptors for glu- genomic effects (39, 50, 106, 156, 157, 177).
cocorticoids are known: the glucocorticoid receptor (GR) Differences between MR and GR are also found in their
and the mineralocorticoid receptor (MR). These receptors distribution throughout the body. While GR is ubiquitously
belong to the superfamily of nuclear receptors (48, 123). expressed in the periphery and in the brain, the distribution of
When inactive, they reside in the cytoplasmic compartment, MR is more localized to specific organs such as the kidney and

1274 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

the heart. In the brain, the distribution of these receptors is also Circadian Rhythms
not uniform (156, 157). GR is almost ubiquitously expressed
in neurons and glial cells, with particularly high density in the The suprachiasmatic nucleus and the clock
hippocampus and in parvocellular neurons of the PVN. In con- gene network
trast, MR localization is restricted to the hippocampus, pre- From the Latin circa diem, circadian means “around a day,”
frontal cortex, and amygdala (3, 10, 198), structures essential and circadian rhythms refer to biological oscillations with a
for learning, memory, and emotional behavior. Significantly, period of approximately 24 h. Circadian rhythms are highly
both receptors are expressed abundantly in areas regulating conserved among living organisms, from cyanobacteria to
the HPA axis, including a number of limbic structures that mammals. Circadian rhythms are characteristic of a num-
regulate the activity of the PVN, such as the hippocampus ber of biochemical, physiological, endocrine, and behavioral
and amygdala, as well as in the anterior pituitary (79). functions, and this allows the organism to anticipate and pre-
pare for predictable environmental changes (i.e., the light/dark
cycle). The result is an optimization of energy expenditure by
Nongenomic effects of glucocorticoids timely coordination of physiological processes.
In addition to their genomic effects, glucocorticoids can also In mammals, circadian rhythms are generated and syn-
modulate cellular activity much more rapidly (i.e., within sec- chronized by a central clock in the suprachiasmatic nucleus
onds to minutes) through nongenomic actions; that is, inde- (SCN) of the ventral hypothalamus (126,135,188). The activ-
pendently of gene expression and de novo synthesis of mRNA ity of the SCN is entrained by the light input received from the
and protein. retina via the retino-hypothalamic tract, and this allows our
Rapid nongenomic effects of glucocorticoids occur physiology to be entrained to solar time (72,136). The molecu-
through a mechanism mediated by G-protein coupled lar mechanism regulating the circadian activity of each single
membrane-associated receptors, with a pharmacological pro- cell within the SCN consists of an oscillating transcriptional
file different from the well known intracellular GR (141,142). network, including the “clock genes” circadian locomotor
Given the rapidity with which glucocorticoids inhibit the output cycles kaput (CLOCK), brain and muscle aryl hydro-
activity of the HPA axis, this effect is likely to be mediated by carbon receptor nuclear translocator-like (BMAL1), period1-
nongenomic mechanisms at both the level of the pituitary 3 (PER1-3), and cryptochrome1-2 (CRY1-2) (20, 61, 104). A
and the brain. For example, corticosterone—administered network of positive and negative transcriptional, translational
peripherally—suppresses rat hippocampal cell firing within and posttranslational feedback loops regulates the rhythmic
20 min (146), while mEPSC frequency increases within min- transcription of these genes, with subsequent rhythmic expres-
utes in CA1 pyramidal cells from rat hippocampal slices sion of their protein products (155). Signaling between the
infused with corticosterone. In the rat, a rapid inhibitory SCN and the periphery involves both hormonal and neuronal
effect of glucocorticoids has also been observed in the mechanisms (112). Rhythmic activity of the SCN is translated
activity of PVN neurons projecting to the median emi- into circadian patterns of gene expression in target tissues,
nence (102, 115, 164). Furthermore, a fast inhibitory effect which in turn determine circadian rhythms in the physiologi-
of corticosterone on CRF-induced ACTH secretion has been cal function of that specific organ.
observed in the rat within 15 min of corticosterone admin- In the SCN, the expression of vasoactive intestinal
istration (75). In addition, a rapid (within 20 min) effect of polypeptide (VIP) is under regulation of the clock machin-
the synthetic glucocorticoid methylprednisolone on corticos- ery. VIP is released with a 24-h cycle, and activates and syn-
terone secretion has also been observed (178). chronizes SCN neuronal activity (5, 120). In rodents, lesion-
Studies from Tasker and colleagues have shown that ing of the SCN results in a loss of physiological circadian
nongenomic inhibitory effects of glucocorticoids in the PVN rhythmicity, including body temperature, locomotor activity,
are mediated by a mechanism involving glucocorticoid- and hormone secretion (1, 17, 182, 200), even when the 24-h
induced endocanabinoid synthesis. Indeed, Di et al. (51) have light-dark cycle is maintained, and the circadian rhythm of
shown that rapid glucocorticoid effects on PVN neuronal locomotor activity can be reinstated in arrhythmic animals by
activity are mediated by the activation of postsynaptic recep- transplantation of an intact SCN (153).
tors and the dendritic synthesis and release of an endogenous In addition to the central master clock in the SCN,
endocanabinoid, which in turn suppresses excitatory synap- clock genes are expressed rhythmically in peripheral tissues,
tic inputs by feeding back onto glutamatergic pre-synaptic including liver, kidney, skeletal muscle, lung, and adrenal
terminals. (16, 70, 71, 187). However, although the molecular dynamics
Evidence for rapid glucocorticoid actions in the brain is of clock genes are the same in the SCN and the peripheral
also supported by behavioral studies. For example, corticos- clocks, only the SCN functions as a self-sustaining pace-
terone has been shown to inhibit male reproductive behav- maker at the whole-tissue level (139, 208, 216); while the
ior as well as medullary neuronal firing in newts (160). SCN can independently generate and maintain its own circa-
Similarly, corticosterone has been shown to have a rapid dian rhythms, peripheral clocks require SCN-dependent drive
effect on aggressive behavior, by modulating glucocorticoid- to maintain synchronicity with the light-dark cycle (139,208).
responsive hypothalamic areas (108). Nevertheless, the existence of peripheral clocks suggests that

Volume 4, July 2014 1275


HPA Axis-Rhythms Comprehensive Physiology

local clock gene networks can generate and regulate the cir- circadian pacemaker that can regulate glucocorticoid synthe-
cadian rhythm of a number of cellular functions, such as daily sis independently from the SCN (144, 175).
variations in metabolic and cardiovascular activity, and hor-
mone secretion. Indeed, disruption of clock gene expression
in the brain and muscle has been associated with a loss of cir-
SCN regulation of HPA axis circadian rhythms
cadian rhythmicity in locomotor activity (127). Furthermore,
ablation of clock genes in the liver leads to a dysfunction Neuro-anatomical tracing studies have identified direct axonal
in glucose and lipid metabolism (109, 110), and disruption projections from the SCN to the PVN, as well as indi-
of clock genes in the pancreas is associated with diabetes rect connections via the dorsomedial hypothalamus (DMH)
mellitus in mice (124). (204, 205). In turn, circadian secretion of CRH regulates cir-
cadian release of ACTH from the anterior pituitary (26, 206).
Interestingly, the expression of StAR in the adrenal cor-
tex, which is dependent on ACTH signaling, is also char-
Circadian Rhythm of Glucocorticoid acterized by a circadian rhythm, as shown by changes in
Secretion both mRNA and protein levels throughout the 24-h cycle
(63, 145, 175, 195), and this in turn regulates circadian syn-
Consistent with other circadian physiological functions, the thesis and release of glucocorticoids (Fig. 2).
circadian rhythm of the HPA axis is primarily regulated by A number of studies have demonstrated a role for vaso-
the central pacemaker located in the SCN (95). The SCN pressin release from the SCN in modulating HPA circadian
regulates the circadian rhythm of glucocorticoids in part by activity. Vasopressin-containing neurons in the dorsal SCN
modulating the release of CRH from neurons in the PVN, and have been shown to be an important component of the SCN
in part via the autonomic nervous system (ANS) through a output (94), and experiments using micro-infusions of vaso-
multisynaptic neural pathway from the SCN to the adrenal pressin and its antagonists in the PVN and the DMH have
gland. Additionally, there is evidence of an intra-adrenal shown that vasopressin released from SCN terminals strongly

(A) (B)
140 Adrenal 400
Plasma
400
Corticosterone (ng/mg)

Corticosterone (ng/mL)
120
300
ACTH (pg/mL)

100 300
200
80 200
100
60
100
40 0
0
5 9 13 17 21 1 5 5 9 13 17 21 1 5
Clock time (h) Clock time (h)

(C) (D)
8 mRNA 1.5
Protein
MC2R mRNA (fold change)
StAR protein (fold change)
StAR mRNA (fold change)

2.0
6 1.25
1.6
4 1

1.2
2 0.75

0.8
0 0.5
5 9 13 17 21 1 5 5 9 13 17 21 1 5
Clock time (h) Clock time (h)

Figure 2 Circadian rhythms in the HPA axis. Rat ACTH (A) and corticosterone (B) profiles
show a clear circadian variation over the 24-h period with peak levels during the active
phase. Steroidogenic acute regulatory protein (StAR), the rate limiting protein in adrenal
glucocorticoid synthesis, is also expressed in the adrenal with a circadian pattern that is
similar to that observed for ACTH and corticosterone (C). In contrast, the expression pattern
of the specific ACTH receptor melanocortin 2 (MC2R) appears to be in antiphase with both
StAR and ACTH (D). Reproduced with permission from (145).

1276 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

inhibits the release of adrenal corticosterone in the rat (93). ANS regulation of the glucocorticoid
Furthermore, a decrease in vasopressin release from these circadian rhythm
SCN terminals in the DMH toward the active dark phase Although a circadian rhythm of ACTH has been described
is necessary for the circadian increase in plasma corticos- in the rat, the circadian variation in corticosterone release
terone (97). The important role of vasopressin in regulating throughout the 24-h cycle is more pronounced than the circa-
low corticosterone levels during the circadian nadir of HPA dian variation in ACTH release. Indeed, the circadian rhythm
axis activity has also been confirmed in a series of experi- in ACTH release is often undetectable (4, 44, 97, 98, 145, 195,
ments in vitro using brain slices in which removal of the SCN 210). This dissociation between ACTH and corticosterone
from the slice results in a loss of circadian rhythm in the during the circadian cycle suggests a diurnal variation in the
spontaneous firing rate of PVN neurons (194). However, as adrenal sensitivity to ACTH, with higher responsiveness dur-
shown in the same study, this rhythm can be reinstated either ing the peak phase of glucocorticoid secretion. Indeed, it has
by using co-cultures of the slice with SCN tissue, or by a been proposed that, in addition to the regulatory effect on
rhythmic (12 h on, 12 h off) perfusion of vasopressin. CRH secretion, the SCN can also regulate adrenal activity
In nocturnal animals, where light entrains the SCN dur- independently from hypothalamic-pituitary drive by modulat-
ing their inactive phase, vasopressin release is important to ing adrenal sensitivity to ACTH. The adrenal gland receives
ensure low circulating levels of corticosterone during the cir- sympathetic innervation via the thoracic splanchnic nerve.
cadian nadir (96). In diurnal species (e.g., man), vasopressin Using transneuronal virus tracing, Buijs and colleagues (19)
release from the SCN is also induced by light, suggesting that have identified direct connections between the adrenal gland
vasopressin has an excitatory effect on HPA axis activity in and neurons located in the intermedio-lateral column of the
diurnal organisms. Indeed, this is in accordance with the well- spinal cord, which receive input from the autonomic division
characterized excitatory effect of vasopressin on its neuronal of the PVN, which in turn is innervated by vasopressin neu-
targets (89, 107). These observations clearly suggest that the rons from the SCN. This is consistent with studies from Jasper
mechanism through which vasopressin exerts an inhibitory and Engeland (87) showing that transection of the splanchnic
or excitatory effect on CRH release in nocturnal or diurnal nerve in the rat increases corticosterone secretion in the morn-
species, respectively, does not involve a direct effect of vaso- ing but has no effect in the evening, suggesting an inhibitory
pressin on CRH neurons, but depends on vasopressin acti- effect of the splanchnic nerve on corticosterone secretion dur-
vation of either an inhibitory or excitatory neuronal pathway ing the nadir phase of the circadian rhythm. Further studies
regulating CRH release in the PVN. Indeed, although there from the same group have also demonstrated a role for the
are direct projections from the SCN to the PVN (204, 205), splanchnic nerve in the regulation of adrenal sensitivity to
there is currently no evidence for a direct connection between ACTH in nonstressed rats (88). The mechanism by which the
fibers from SCN vasopressin neurons and CRH neurons in splanchnic nerve regulates adrenal sensitivity to ACTH is not
the PVN (18, 199). fully understood. A study in which the adrenal responsiveness
A detailed anatomical scheme of the SCN neuronal con- to different doses of exogenous ACTH was investigated (195)
nections in both nocturnal and diurnal species has been pro- has shown that the decrease in corticosterone levels induced
posed by Kalsbeek and colleagues by comparing the effect by splanchnic nerve transection during the peak phase of hor-
of vasopressin on CRH release in the nocturnal rat and in mone secretion is associated with a decrease in the levels of
the diurnal Sudanian grass rat (Arvicanthis ansorgei) (95). In adrenal cAMP. Since ACTH induces glucocorticoid synthe-
accordance with their model, vasopressin is released from the sis via the cAMP/PKA signaling pathway, these data suggest
SCN during the day in both nocturnal and diurnal species. In that splanchnic nerve activity regulates adrenal sensitivity to
nocturnal species, vasopressin released from the SCN will ACTH by affecting intracellular signaling pathways involved
activate gamma-aminobutyric acid (GABA)ergic interneu- in glucocorticoid synthesis. Via a signaling pathway involv-
rons within the PVN and the DMH, which in turn will directly ing cAMP, PKA, and CREB, ACTH induces transcriptional
or indirectly (via the DMH) inhibit CRH release during the activation of several steroidogenic genes, including StAR.
circadian nadir. In contrast, in the diurnal species, vasopressin However, in this study, the circadian rhythm of StAR was not
release from the SCN will activate glutamatergic interneu- affected by lesioning the splanchnic nerve, suggesting that the
rons in the PVN and DMH, which in turn will stimulate effects of splanchnic activation at the level of adrenal sensitiv-
CRH release. Although the importance of SCN-secreted vaso- ity to ACTH may be modulated by a nongenomic mechanism,
pressin for inhibition of PVN activity has been well described such as phosphorylation of StAR.
in the rat, a neurotransmitter responsible for SCN stimula-
tory effect on the HPA axis during the circadian peak has not
been clearly identified. A number of studies have also sug-
gested that a stimulatory signal from the SCN via the release Circadian clock genes in the adrenal gland
of VIP modulates the evening rise in HPA activity (5, 120). regulate rhythmic glucocorticoid synthesis
Furthermore, a role for neuromedin U in regulating the stim- In addition to the SCN, key components of the HPA axis,
ulatory effect of the SCN on HPA axis activity has also been including the PVN, pituitary, and adrenal cortex, express
proposed (64). circadian clock genes (63, 81, 95, 143, 144). Indeed, using

Volume 4, July 2014 1277


HPA Axis-Rhythms Comprehensive Physiology

microarray and qPCR techniques to analyze circadian gene The mechanisms underlying the regulation of adrenal
expression in the mouse adrenal gland, Oster and col- steroidogeneis by adrenal clock genes have recently been elu-
leagues have recently shown that approximately 5% of the cidated using Bmal1 adrenal knockdown mice (175). This
whole genome—including several canonical clock genes— study has shown that the circadian expression of StAR
demonstrates rhythmic expression (143). Evidence for the appears to be directly regulated by the Clock:Bmal1 com-
involvement of adrenal clock genes in circadian glucocor- plex at the level of the StAR promoter in the adrenal gland.
ticoid synthesis has been shown in vivo using mice defec- Bmal1 adrenal knockdown mice display a flattened circadian
tive in components of the circadian clock system. In fact, rhythm of StAR protein expression, and although the circa-
studies in clock gene knockout mice have shown that dis- dian rhythm of corticosterone is maintained in these animals
ruption of the circadian clock in these animals is associated when they are kept under a normal light-dark cycle, corti-
with a disruption of the HPA circadian rhythm. Dallmann and costerone rhythmicity is lost in mutant mice maintained in
colleagues (46) have shown that, while both Per1 KO mice constant darkness. This suggests that, while in normal light
and Per2 KO mice have markedly elevated levels of circu- conditions adrenal circadian rhythmicity is primarily under
lating glucocorticoids, only Per1 KO mice lack a detectable the control of the SCN via both hypothalamic-pituitary and
circadian hormone rhythm. In agreement with this study, a splanchnic nerve regulation, the adrenal clock pathway may
loss of circadian rhythm of corticosterone has been found in become important for regulation of steroidogenic genes when
Per2/Cry1−/−double mutant mice, whereas circadian rhyth- the central circadian input is disrupted.
micity of ACTH is maintained in these mice (144). Interest- A more recent study has shown that in addition to StAR,
ingly, in these animals, a number of clock genes, including several of its transcriptional regulators are also rhythmi-
Bmal1, Per1, and Per3, which are all rhythmically expressed cally expressed in the adrenal gland (145), and this includes
in the wild-type animals, lack transcriptional circadian rhyth- positive regulators such as the nuclear receptors steroido-
micity. The same authors have shown that transplantation of genic factor 1 (SF-1) (189) and nerve Growth factor IB
wild-type adrenal glands restores corticosterone rhythmic- (NGFIB, also known as Nur77 or NR4A1) (176), as well the
ity, whereas transplanting adrenal glands from arrhythmic negative regulator nuclear receptor dose-sensitive sex rever-
Per2/Cry1 double mutant mice to wild-type adrenalectomized sal, adrenal hypoplasia congenita determining region on the
mice dampens the corticosterone circadian rhythm. Further- X-chromosome-1 (DAX1) (217). Consistent with their role in
more, the circadian rhythmicity of clock genes in the adrenal regulating the expression of adrenal StAR, SF-1 and Nur77
is independent of circadian rhythmicity of ACTH, as has been mRNA and protein levels are high during the hormonal cir-
shown in a recent study in which circadian rhythms of clock cadian peak, whereas levels of expression of DAX1 are high
genes such as Per1, Per2, and Bmal1 are maintained in rats during the circadian nadir (145). Whether circadian variation
after hypophysectomy (58). Taken together, these observa- in StAR’s transcriptional regulators is important for the circa-
tions suggest that, although the central pacemaker in the SCN dian corticosterone rhythm, via regulation of the StAR circa-
controls peripheral clock gene rhythmicity, clock genes in the dian rhythm, is not known. However, Park and colleagues have
adrenal may have an important role in regulating circadian shown that disruption of the corticosterone circadian rhythm
rhythms of glucocorticoid synthesis. in rats kept in constant light is associated with a disrupted
Several rhythmically expressed genes in the adrenal circadian rhythm of StAR and its transcriptional regulators
gland encode proteins involved in steroidogenic processes, (145). Whether transcriptional activation of StAR’s regula-
including proteins involved in cholesterol biosynthesis and tors is under control of the clock gene machinery, as is the
transport (e.g., StAR), and proteins implicated in ACTH case for StAR, is also not currently known. Taken together,
receptor signaling [e.g., MC2R and the melanocortin 2 recep- these data suggest that, while the SCN is indispensable for
tor accessory protein (MRAP)] (144, 145, 175). Park and maintaining a circadian rhythm of glucocorticoid secretion,
colleagues have found that the pattern of StAR expression the adrenal clock provides an additional level of control by
is consistent with the pattern of ACTH and corticosterone regulating the steroidogenic pathway.
secretion; that is, StAR mRNA and protein levels are max-
imal during the active phase. Interestingly, the pattern of
expression of MC2R mRNA appears to be in antiphase with
both StAR expression and ACTH and corticosterone secretion Ultradian Rhythm of Glucocorticoid
(Fig. 2D). A recent study has shown that, while the ACTH Secretion
circadian rhythm is unchanged in Bmal1 deficient mice, cir-
cadian variations of corticosterone are reduced, with lower As shown for other neuroendocrine systems that sig-
corticosterone levels during the peak phase (111). Interest- nal through the hypophysial portal system, such as the
ingly, the effect of Bmal1 deficiency on the corticosterone gonadotropic hormone and growth hormone pathways, the
rhythm in these animals is associated with downregulation in basal activity of the HPA axis is characterized by pulsatile
the expression of several key genes involved in cholesterol activity. The circadian variation in glucocorticoid levels over
trafficking, including StAR, while the expression of MC2R is the 24-h cycle is not made up of a smooth change in hor-
elevated. mone levels, but is in fact characterized by a rapid ultradian,

1278 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

pulsatile pattern of hormone secretion, with a periodicity of other components of the HPA axis. Indeed, a small number
approximately one hour (Fig. 1B). The ultradian glucocorti- of studies, both in vitro and in vivo, have reported episodic
coid rhythm has been reported in numerous species, including release of CRH from macaque hypothalamic explants (131),
rat (86,214), rhesus monkey (76,192), sheep (59), and human rapid fluctuations in CRH levels in the median eminence of
(73, 113, 207). freely behaving rats (83,85), and pulsatile patterns of CRH and
The ultradian rhythm of corticosterone in the rat was first AVP in the hypophysial-portal circulation of conscious sheeps
described by Jasper and Engeland using intra-adrenal micro- (24, 55). In addition to this, ultradian pulses of ACTH have
dialysis in nonstressed freely behaving animals (86). In addi- been observed in the rat (25,26,29), sheep (8), and human (73).
tion to the hourly corticosterone pulse frequency, they also Observations of pulsatile CRH have led to a general
observed that both the ultradian frequency and amplitude of acceptance that a ‘hypothalamic “pulse generator”’ drives the
the pulses are modulated in a circadian manner (87). ACTH and glucocorticoid ultradian rhythm. However, a num-
The pattern of corticosterone secretion has also been ber of findings are in contrast to this hypothesis. Significantly,
investigated in blood plasma. An automated blood-sampling there is a mismatch between the frequency of CRH pulses and
(ABS) system developed by Clark et al. (37) enables the the frequency of ACTH and corticosterone oscillations. In the
remote collection of small blood samples at a high frequency rat, CRH pulse frequency is higher (∼3 pulses/h) (83) than
(e.g., every 5-10 min) over extended periods of time (214). the near-hourly oscillation in ACTH (26) and glucocorticoids
This system provides a powerful tool to study glucocorticoid (214) (Fig. 4). Furthermore, studies in the sheep have shown
levels in “unstressed” rats over the whole 24-h cycle, and that pituitary-adrenal ultradian activity is maintained follow-
has been used to show that corticosterone levels in the blood ing hypothalamic disconnection from the pituitary (56), sug-
are also pulsatile with an ultradian frequency. Analysis of gesting that a supra-pituitary pulsatile drive is not required
pulse characteristics using the PULSAR algorithm (130) has for pulsatility in ACTH and glucocorticoids.
shown that the pulse amplitude varies in a circadian manner, The pituitary-adrenal system is characterized by both
with low amplitudes in the morning and higher amplitudes in a positive feed-forward pathway whereby pituitary ACTH
the evening (213). drives glucocorticoid release from the adrenal cortex, and a
negative feedback mechanism through which adrenal glu-
cocorticoids feed back on the anterior pituitary to inhibit
Ultradian rhythms of the HPA axis ACTH release. A short time delay in the pituitary-adrenal
There is good evidence that the circadian rhythm of glucocor- feed-forward pathway has been observed both in humans
ticoids is regulated by the SCN. However, the mechanisms and in the rat, where each pulse of ACTH is followed by
generating the ultradian pulsatile dynamics of the system a delayed response in cortisol or corticosterone, respectively
remain poorly understood. In rats with SCN lesions, the cir- (27,73) (Fig. 5). Furthermore, a rapid inhibitory effect of glu-
cadian rhythm of corticosterone is disrupted, but the ultradian cocorticoids on CRH-induced ACTH secretion from the ante-
pulsatile pattern of corticosterone is maintained (200), which rior pituitary has been shown (75, 90, 92, 122, 161-163, 209).
suggests that the origin of the ultradian rhythm of corticos- The observation of this fast inhibitory feedback within the
terone is generated independently of the SCN (Fig. 3). pituitary-adrenal system has led to the hypothesis that this
A number of studies have shown that, in addition to process could provide a potential mechanism within the sys-
endogenous glucocorticoids, an ultradian pattern underlies tem for generating oscillatory dynamics.

(A) (B)
100
Corticosterone (ng/mL)

80

60

40

20

0
0900 1500 2100 0300 0900 0900 1500 2100 0300 0900
Clock time (h) Clock time (h)

Figure 3 Ultradian corticosterone pulsatility is maintained following lesioning of the SCN. (A)
Plasma corticosterone levels in a control (sham lesion) rat. (B) Plasma corticosterone levels in a
rat with SCN lesion. Corticosterone was measured in blood samples collected at 10 min intervals
from freely behaving male Sprague-Dawley rats using an automated blood sampling system.
Gray curves indicate circadian trend. Shaded regions indicate the dark phase. Reproduced with
permission from (200).

Volume 4, July 2014 1279


HPA Axis-Rhythms Comprehensive Physiology

(A) 40 800
CRH-41 (pg/5 min fraction) 20

Plasma ACTH (pg/mL)

Serum cortisol (nmol/l)


ACTH
15 30 600 Cortisol

10 20 400

5
10 200
0
1700 1800 1900 2000
Clock time (h) 0 0
(B) 200 0200 0500 0800 1100 1400 1700
Clock time (h)
Plasma ACTH (pg/mL)

160
Figure 5 Human cortisol and ACTH dynamics. Ultradian ACTH and
cortisol oscillations in a healthy man are tightly correlated with ACTH
120 leading cortisol. ACTH and cortisol were measured in blood samples
collected at 10 min intervals using an automated blood sampling sys-
tem. Light was off between 2300 and 0700. Adapted and reproduced
80
with permission from (73).

40
oscillations with a physiological ultradian frequency, even
0 under conditions of constant CRH drive to the anterior pitu-
1000 1100 1200 1300 1400 1500 itary. Analysis of this model has provided a number of the-
Clock time (h) oretical predictions (Fig. 6). First, the model predicts that
(C) 120
oscillations in ACTH and glucocorticoids can be generated
for “intermediate” constant levels of CRH, and that these
Corticosterone (ng/mL)

100
oscillations occur at the same frequency as endogenous cor-
80 ticosterone oscillations observed in the rat during the peak
phase of the circadian cycle (Fig. 6, A and B). Moreover,
60
the model also suggests that these glucocorticoid oscillations
40 become “damped” for higher (or lower) levels of CRH (Fig. 6,
A and C). These predictions are consistent with experimental
20 observations that ACTH and corticosterone pulse amplitude
0
increases during the early dark phase are accompanied by
1500 1700 1900 2100 increased mean levels of CRH in the median eminence (84),
Clock time (h) and with the loss of pulsatile corticosterone secretion that
occurs both at the circadian nadir of HPA axis activity and
Figure 4 An ultradian pattern underlies all the components of the following an acute stress response (214), when the levels of
HPA axis. (A) Pulsatile release of CRH in the rat median eminence.
CRH was measured in perfusate from push-pull cannulas implanted in CRH are respectively lower and higher than during the cir-
the rat median eminence; samples were collected at 5 min intervals cadian peak. The third key prediction of this mathematical
from an unanesthetized male rat under basal conditions. Reproduced model is that glucocorticoid oscillations induced by constant
with permission from (84). (B) ACTH levels in rat plasma samples col-
lected every 2 min from the jugular vein via an indwelling cannula. CRH stimulation are driven by oscillations in ACTH, with
Reproduced with permission from (27). (C) Corticosterone levels in rat a phase shift between ACTH and glucocorticoids. This is
blood samples collected every 5 min using an automated blood sam- again consistent with experimental data showing that ultra-
pling system. Reproduced with permission from (201). Note that in the
rat, CRH pulse frequency is higher (∼3 pulses/h) than the near-hourly dian ACTH oscillations have been observed in the rat (26,27)
oscillation in ACTH and corticosterone. and that pulsatile ACTH is a critical factor in regulating pul-
satile corticosterone secretion from the adrenal cortex (180).
Consistent with both mathematical and experimental data,
high-frequency blood sampling in humans under basal, non-
The origin of the glucocorticoid stressed, conditions has revealed a close temporal relationship
ultradian rhythm between pulses of ACTH and cortisol, with a short phase lag
The interaction between the pituitary corticotrophs and the between the two hormones (73) (Fig. 5).
adrenal fasciculata cells has been modeled mathematically The hypothesis that the sub-hypothalamic pituitary-
utilizing differential equations that incorporate the rapid glu- adrenal system can function as an ultradian hormone oscil-
cocorticoid inhibition of ACTH secretion (202). Numerical lator has been tested in vivo by studying the dynamics of
analysis of the model suggests that the pituitary-adrenal sys- corticosterone responses to different levels of constant CRH
tem can support self-sustained ACTH and glucocorticoid stimulation in conscious freely behaving male rats (201). In

1280 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

Oscillation period (min)


(A)
45 60 75

60 (B) ACTH
2.33 2
CORT

CORT (AU)
ACTH (AU)
50 Steady state

40 C 0 0
0 1 2 3 4 5
CRH (AU)

Time (h)
30
Oscillation
B (C) 2.33 2 ACTH

CORT (AU)
ACTH (AU)
20 CORT

10
0 0
0 1 2 3 4 5
0 Time (h)
0 5 10 15 20
Adrenal delay (min)

Figure 6 Mathematical modeling predictions of the pituitary-adrenal response to different levels


of constant CRH drive. (A) Computed two-parameter bifurcation diagram shows that different
combinations of constant CRH drive and adrenal delay result in qualitatively different dynamic
responses from the pituitary-adrenal system. On one side of the transition curve, the pituitary-
adrenal system responds with oscillations in ACTH and glucocorticoids (CORT), despite the fact
that the CRH drive is constant (point B). On the other side of the transition curve, the pituitary-
adrenal system responds with steady state levels in ACTH and CORT (point C). (B) Model predictions
for ACTH (grey) and CORT (black) corresponding to point B in panel (A). (C) Numerical simulations
for ACTH (grey) and CORT (black) corresponding to point C in panel (A). Model predictions are
shown for a 5-h period after transient dynamics have decayed. AU, arbitrary units. Reproduced
with permission from (202).

the male Sprague-Dawley rat, the HPA circadian cycle is char- provides a mechanism for generating ultradian oscillations in
acterized by a distinct and prolonged nadir phase (7 am-1 pm), ACTH and glucocorticoid hormone secretion has been sup-
when endogenous CRH secretion is minimal (84) and there is ported by both mathematical modeling and experimental data
no detectable pulsatile secretion of ACTH or corticosterone and these findings provide good evidence for an oscillating
(28,214). This experimental model allows manipulation of the mechanism outside the central nervous system.
system by controlling the level of CRH and simultaneously
measuring the levels of ACTH and corticosterone, without
interference from endogenous HPA hormone release. Con- Adrenal regulation of glucocorticoid pulsatility
stant infusions of CRH (during the circadian nadir) have been Glucocorticoid hormones are lipophilic and therefore can-
shown to induce different temporal patterns of corticosterone not be simply packaged in readily releasable vesicles within
secretion that are dependent on the level of CRH. In keep- steroidogenic cells in the adrenal cortex, but need to be
ing with the predictions of the mathematical model (202), newly synthesized in response to ACTH. Given that pulsatile
a constant infusion of CRH induces sustained ultradian cor- secretion of glucocorticoids is generated by pulses of ACTH,
ticosterone oscillations that persist throughout the infusion as shown by both mathematical modeling and experimental
period, with pulse amplitude remaining relatively constant work (201, 202), it is clear that within the adrenal there must
throughout the infusion (Fig. 7A). Furthermore, there is a be a remarkably responsive steroid synthesis mechanism that
phase shift between ACTH and corticosterone, with ACTH is able to respond rapidly to each pulse of ACTH. Indeed,
oscillations preceding corticosterone oscillations, also pre- a recent study in the rat has shown that pulsatile ACTH
dicted by the mathematical model (Fig. 7, B and C). This is necessary for optimal release of corticosterone (180); in
time delay between ACTH and corticosterone pulses reflects rats in which endogenous HPA activity has been suppressed
the time taken for de novo biosynthesis and release of corticos- by the addition of a synthetic glucocorticoid (methylpred-
terone from the adrenal cortex upon stimulation with ACTH nisolone, MP) to the drinking water, pulsatile infusion of
(186). Interestingly, this oscillatory response to CRH is dose ACTH results in a normal pattern of pulsatile corticosterone
dependent, and becomes disrupted for higher levels of CRH. secretion (Fig. 8A), whereas an identical dose of ACTH
In summary, the hypothesis that a systems-level dynamic infused at a constant rate has no effect on the adrenal cor-
balance between positive feed-forward and negative feedback ticosterone secretion.

Volume 4, July 2014 1281


HPA Axis-Rhythms Comprehensive Physiology

(A) 150 (B) 80 140 (C) 100 160


ACTH ACTH

Corticosterone (ng/mL)

Corticosterone (ng/mL)
120 140
Corticosterone (ng/mL) CORT CORT
80 120

ACTH (pg/mL)

ACTH (pg/mL)
100
100 60
100
80
60 80
60
60
50 40
40 40 40
20 20
0 20 0 20 0
0600 0800 1000 1200 0700 0800 0900 0 10 20
Clock time (h) Clock time (h) Time (min)

Figure 7 Constant infusion of CRH induces pulsatile secretion of ACTH and corticosterone. (A) Individual corticosterone
response to constant CRH infusion. In line with the modeling hypothesis, constant infusion of CRH (0.5 μg/h) induces ultradian
corticosterone oscillations that persist throughout the infusion period. This oscillatory response is characterized by an initial
rapid increase in corticosterone within approximately 15 min following the onset of the CRH infusion, reaching peak levels by
approximately 30 min and returning to low levels by approximately 80 min. Following the initial pulse, the amplitude of the
oscillation is relatively constant throughout the infusion. Samples were collected every 5 min from a freely behaving male rat
using an automated blood sampling system. Grey bar indicates the period of infusion. (B) ACTH and corticosterone responses to
constant CRH infusion. Samples were collected manually at 20 min intervals throughout the infusion via an indwelling cannula
implanted in the jugular vein. In agreement with the modeling predictions, CRH induces ACTH and corticosterone oscillations
that persist throughout the infusion period. (C) Phase-shifted ACTH and corticosterone response to constant CRH infusion
(0.5 μg/h) over the initial activation phase (0-25 min) of the oscillation. This phase shift between ACTH and corticosterone
presumably reflects the time taken for de novo biosynthesis and release of corticosterone in the adrenal cortex. Reproduced with
permission from (201).

To better understand the mechanism underlying the in vivo in vitro (190, 191). A recent study has shown that a pulse of
generation of pulsatile corticosterone secretion from adrenal ACTH also induces pulsatile transcription of the gene encod-
fasciculata cells within the adrenal cortex, the dynamics of ing for MRAP (119), a protein that is crucial in regulating
steroidogenic gene transcription in relationship to the dynam- the level and activity of MC2R at the cell surface, and thus
ics of corticosterone secretion has been investigated over a the cell’s responsiveness to ACTH (132) (Fig. 8C). Taken
detailed time-course following a single ultradian pulse of together, these studies clearly suggest that pulsatile secretion
ACTH (179). In the rat, pulsatile release of corticosterone, of corticosterone is associated with pulsatile transcription of
induced by intravenous (IV) administration of a small ACTH steroidogenic genes in the adrenal gland.
pulse, is associated with pulsatile transcription of steroido- Although corticosterone levels peak within 5 min of an
genic genes, including StAR (118, 186) (Fig. 8B). This has ACTH pulse (given IV), hnRNA StAR levels do not peak until
been shown by measuring rapid changes in the levels of het- 15 min after the ACTH pulse. The fact that the peak of the
eronuclear RNA (hnRNA), which is a reliable marker of gene secretory event precedes the transcription peak of the steroido-
transcription. Specifically, StAR hnRNA levels peak at 15 min genic enzyme is consistent with evidence that nongenomic
after ACTH administration, and return to basal levels within posttranscriptional and posttranslational changes in the StAR
30 min. A similar dynamic pattern of transcription has been protein regulate the rapid steroidogenic response. Indeed,
observed for CYP11A, the gene encoding for cholesterol side- rapid activation of proteins involved in steroidogenesis by
chain cleavage cytochrome P450 (P450scc) (36), which catal- phosphorylation, such as phosphorylation of StAR on Ser194
yses the cleavage of the cholesterol side chain to produce mediated by PKA, plays a role in the immediate initiation
pregnenolone in the inner mitochondrial membrane. In keep- of steroidogenesis and glucocorticoid secretion upon stimu-
ing with these findings, Spiga and colleagues (179) have also lation of the adrenal by ACTH (31, 133, 134, 148). Therefore,
shown that activation of CREB and its transcriptional regu- it is possible that the pulsatile transcription of StAR following
lator TORC2 (transducer of regulated CREB activity 2, also each pulse of ACTH may be acting as mechanism for replen-
known as CRTC2) also occurs in a highly dynamic pattern ishing depleted intracellular stores of StAR (that have been
following pulsatile ACTH stimulation: PKA-mediated activa- used to generate a pulse of corticosterone) prior to subsequent
tion of CREB and TORC2, by phosphorylation and dephos- ACTH pulses. Furthermore, these observations are consistent
phorylation respectively, occurs within 5 min of exposure to with previous data showing that only newly synthesized StAR
a pulse of ACTH, supporting an involvement of these pro- is involved in posttranslational events mediating the rapid
teins in the rapid (within 15 min) transcription of StAR. An synthesis of corticosterone (11). Whether nongenomic events
involvement of TORC2 in the CREB-mediated transcriptional regulating rapid steroidogenesis are also characterized by a
regulation of StAR in adrenalcortical cells has been shown pulsatile pattern of activity is currently under investigation.

1282 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

(A) 100

Corticosterone (ng/mL)
75

50

25

0700 1300 1900 0100 0700


Clock time (h)

(B) 2.5 (C) 10

2.0 8
Fold induction hnRNA

Fold induction hnRNA


(MRAP/GAPDH)
(StAR/GAPDH)

1.5 6

1.0 4

0.5 2

0 0
0 5 15 30 60 0 5 15 30 60
Time (min) Time (min)

Figure 8 Effect of pulsatile ACTH administration on plasma corticosterone levels and StAR and
MRAP primary transcript levels. (A) Pulsatile corticosterone secretion in rats chronically treated with
methylprednisolone (MP) and infused with pulsatile ACTH (4 ng/h, 5 min pulse/h). ACTH infusion
started at 10 am and blood samples were collected every 10 min using an automated blood sampling
system. Data are the mean of 9 different rats. Data reproduced with permission from (180). (B and
C). Administration of a small dose of ACTH (4ng/rat; i.v.) induces a rapid but transient increase in the
primary transcript of StAR (B) and MRAP (C), suggesting that pulsatile expression of steroidogenic genes
is important for the ultradian rhythm of corticosterone secretion. Transcriptional response of StAR and
MRAP was measured by RT-qPCR using specific primers targeting the gene primary transcript. Data in
(B) reproduced with permission from (179). Data in (C) reproduced with permission from (119).

Glucocorticoid Rhythms in Health Recently, studies using frequent blood sampling to investigate
corticosterone pulsatility in the rat have shown a profound
and Disease difference in the pulsatile profile of corticosterone release
Over the last 20 years, we have used our ABS system to define over the 24-h cycle between male and female rats (169-172).
corticosterone pulsatility in freely behaving rats in their home Compared to male rats, basal corticosterone secretion is
cages. These studies have revealed that both the circadian enhanced in female rats (169) (Fig. 9, A and B), and this is
and the ultradian pattern shows marked sexual diergism and due to: (i) an increase in the total number of pulses over the
genetic variation. Furthermore, corticosterone rhythms are 24-h cycle (28 pulses for females vs. 10 pulses for males);
remarkably plastic, with changes seen both in physiological (ii) an increase in pulse magnitude (approximately 60 ng of
conditions, such as during lactation and ageing, and in patho- corticosterone/pulse for females vs. 24 ng/pulse for males);
logical conditions, particularly stress-related disorders. and (iii) a greater number of pulses over the 24h period.
Gonadal steroids have major effects on the pulsatile pat-
tern of corticosterone release in the rat. Following gonadec-
Physiological changes: Gonadal steroids tomy, male rats have increased overall corticosterone secre-
A marked difference in the levels of basal corticosterone tion similar to that observed in female rats, characterized
secretion between male and female rodents is well recognized. by an increased number of pulses, pulse amplitude, and

Volume 4, July 2014 1283


HPA Axis-Rhythms Comprehensive Physiology

120

Corticosterone (ng/mL)
(A) (B)
100
80
60
40
20
0
0700 1300 1900 0100 0700 0700 1300 1900 0100 0700
Clock time (h) Clock time (h)
160
Corticosterone (ng/mL)

(C) (D)
120

80

40

0
0500 1100 1700 2300 0500 0500 1100 1700 2300 0500
Clock time (h) Clock time (h)
120 (E)
Corticosterone (ng/mL)

(F)
100
80
60
40
20
0
1800 0000 0600 1200 1800 1800 0000 0600 1200 1800
Clock time (h) Clock time (h)

Figure 9 Changes in ultradian glucocorticoid dynamics in different physiological and


pathological states. (A and B) Ultradian corticosterone oscillations in a female (A) and male
(B) Sprague-Dawley rat. Reproduced with permission from (169) (C and D). Ultradian corti-
costerone oscillations in juvenile (C) and adults (D) rats (unpublished observations). (E and F)
Ultradian corticosterone oscillations in a control male PVG rat (E) and a male PVG rat with
active immune-mediated adjuvant-induced arthritis (F). Reproduced with permission from
(212). In all experiments, corticosterone was measured in blood samples collected at 10 min
intervals from freely behaving rats. Shaded regions indicate the dark phase.

pulse frequency. On the other hand, ovariectomized females these pulse characteristics are comparable to those observed
have reduced overall corticosterone levels similar to those in normal adult male rats (171). Consistent with this, changes
observed in male rats, with a reduction in the number of in the adult pulsatile pattern of corticosterone secretion are
pulses, and decreased pulse amplitude and frequency (169). observed in male rats exposed to neonatal feminization (172):
To better understand whether these changes are due to a here, rats deprived of pre and postnatal testosterone activity
direct effect of gonadal hormone withdrawal, further stud- by exposure to the antiandrogen flutamide and the aromatase
ies have been carried out to investigate the effect of androgen inhibitor 1,4,6-androstatriene-3,17-dione, respectively, show
or oestrogen replacement on corticosterone secretion in the increased basal corticosterone secretion characterized by an
gonadectomized male and female, respectively (170): andro- increase in the number, frequency, and amplitude of corti-
gen replacement reverses the increase in corticosterone pulse costerone pulses. Interestingly, the amplitude of the corticos-
frequency and amplitude induced by castration in male rats; terone pulses of prenatal testosterone-deprived rats is similar
similarly, oestradiol reverses the changes in corticosterone to those observed in intact females and castrated males (169).
secretory activity in ovariectomized female rats. The mechanisms underlying the effects of gonadal
Neonatal masculinization or feminization also affects steroids on the ultradian rhythm of glucocorticoids are not
basal corticosterone secretion patterns in adult life in female clear. There is evidence suggesting that androgens and estro-
and male rats, respectively (171,172). Exposure of the female gens can affect HPA axis activity at the level of the adrenal
neonate to a single injection of testosterone within 24 h of birth gland, anterior pituitary and PVN [reviewed in (67)]. This is
results, in adult animals, in a reduction in the number of cor- supported by an overlap in the expression of gonadal hormone
ticosterone pulses, as well as pulse amplitude and frequency, receptor and glucocorticoid hormone receptors in key areas
over the 24-h period, compared to untreated adult females, and regulating the activity of the HPA axis (67).

1284 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

Differences in female corticosterone levels across the Physiological changes: Genetic background
estrus cycle have been observed. During the diestrous phase,
Changes in corticosterone pulsatility patterns related to
corticosterone levels in female rats are similar to those
genetic background have also been observed. Ultradian corti-
observed in male rats; in contrast, during the proestrous phase,
costerone rhythms have been investigated in a number of rat
when the concentrations of progesterone and estradiol are ele-
strains, including Wistar, Sprague Dawley, Lewis, and Fisher
vated, corticosterone levels are increased (34, 45), and this
344. Although they all exhibit a pulsatile pattern of corti-
is paralleled by an increase in CRH mRNA expression in
costerone release, there are significant differences between
the PVN (82). Consistent with this, inhibition of HPA axis
strains (213). Lewis rats have a clear circadian rhythm, as
activity by androgens is paralleled by a reduction in CRF-
seen in female and male Sprague Dawley and Wistar rats, due
immunoreactivity in the PVN (15). Gonadal steroid hormones
to changes in pulse amplitude over the 24-h cycle, which is
can also regulate basal glucocorticoid secretion by affecting
greater in the evening than in the early morning. In contrast,
GR signaling. Indeed, estrogens regulate the transcriptional
female Fischer rats have higher mean circulating corticos-
of a number of proteins involved in GR activity, such as the
terone concentrations and lack any discernible circadian vari-
chaperone protein FKBP51 (78) and the nuclear GR coacti-
ation, and this is due to increased pulse amplitude in the morn-
vator SRC1 (22). Taken together, these studies suggest that
ing, which is similar to pulse amplitude in the evening. The
sex steroids can affect the glucocorticoid ultradian rhythm by
mechanism underlying the different pattern of corticosterone
modulating both hypothalamic CRH drive and GR-mediated
secretion between the two strains is not known. Given that
glucocorticoid negative feedback.
female Fisher rats maintain high-pulse amplitudes throughout
the morning, it has been proposed that insensitivity to corti-
costerone feedback, mediated by activation of both GR and
Physiological changes: Ageing
MR, might be responsible for the lack of circadian rhythm.
Studies on ageing rats have revealed marked changes in the Furthermore, in light of the role of splanchnic innervation of
pattern of corticosterone pulsatility across their life cycle the adrenal in the regulation of circadian adrenal responsive-
(117) (Fig. 9, C and D). In juvenile and adults rats, marked ness to ACTH (195), the differential dynamics in pulsatile
changes in pulse amplitude over the 24-h cycle have been secretion between the two strains could also be due to differ-
observed, with higher amplitude pulses during the peak phase, ences in sympathetic regulation of adrenal sensitivity. Inter-
resulting in the well defined circadian rhythm of corticos- estingly, Lewis rats are known to be susceptible to a range of
terone. In elderly (12-month-old) rats, however, there is a inflammatory conditions, including streptococcal cell wall-
loss of circadian rhythmicity predominantly due to a decrease induced arthritis (183, 184), whereas the Fisher rat does not
in pulse amplitude during the peak phase, but also due to develop any of these conditions. This suggests that differences
an overall loss of ultradian pulsatility throughout the 24 h. in disease susceptibility may be linked to differences in the
These observations are important and suggest a potential link dynamics of corticosterone secretion.
between disrupted corticosterone pulsatility and age-related
disorders.
Pathological changes: Chronic stress
Chronic inflammation is a model of chronic stress that has
Physiological changes: Reproduction
high clinical relevance. In particular, a model of chronic
Changes in corticosterone rhythms have also been observed in inflammatory disease that has been extensively studied in the
female rats over the course of their reproductive cycle (211). rat is Mycobacterium-adjuvant-induced arthritis. Increased
Circadian and ultradian rhythms of corticosterone have been circulating corticosterone and ACTH concentrations, and loss
studied in virgin rats, rats on day 9 of lactation, weaned dams of the normal circadian rhythm of HPA activity, have been
2 days after pups removal, and postlactating dams 13 days observed in Piebald-Viral-Glaxo (PVG) rats infected with the
after pups removal. During lactation, although there is a sig- Mycobacterium-adjuvant (212) (Fig. 9, E and F). While con-
nificant circadian variation in corticosterone release, this is trol PVG rats demonstrate the expected circadian and ultra-
associated with a flattening of the rhythm—due to a decrease dian patterns of corticosterone release, significant changes in
in the evening peak levels and an increase in the number of cor- the dynamics of corticosterone secretion occur both prior to
ticosterone pulses throughout the 24 h. Very marked changes the development of arthritis (presymptomatic rats sampled at
in corticosterone release are also observed in rats 2 days after 6 days after injection of the mycobacterium) and following
experimental weaning. In this group, corticosterone levels are the development of hind-joint inflammation (13 days after
significantly suppressed throughout the 24-h period, with the injection). In presymptomatic rats, although a diurnal pattern
number of pulses and pulse amplitude significantly lower than of corticosterone secretion is still observed, there are changes
any other group. Interestingly, these effects are reversible, as in the characteristics of the pulses. While no changes in the
no differences in either circadian or ultradian rhythms of cor- number of pulses or their amplitude have been observed across
ticosterone are observed between 13 days postlactating dams the 24-h cycle, in presymptomatic rats the average length
and virgin rats. of time during which hormone levels rise from baseline to

Volume 4, July 2014 1285


HPA Axis-Rhythms Comprehensive Physiology

peak is decreased. Specifically, within each pulse, corticos- of the organism. The effect of neonatal programming on cor-
terone levels rise to peak concentrations more quickly than in ticosterone pulsatility in the rat has been investigated using a
control rats. During the symptomatic period, rats show dra- model of early life infection (174). In rats exposed to endo-
matic changes in pulsatility, with an almost doubled number toxin (Salmonella enteritidis) in neonatal age (days 3 and 5
of pulses throughout the 24-h cycle related to continued pul- postpartum), basal levels of corticosterone are higher both
satility during the normally quiescent lights-on period. Inter- during the nadir and peak phase of the circadian cycle in
estingly, neither the amplitude or the duration of the pulses adult life. This is due to an increase in both the number and
are different from control rats, indicating that the increase in amplitude of corticosterone pulses throughout the 24-h cycle.
circulating hormone is solely due to this increased number of
pulses. It is important to note that in rats exposed to adjuvant-
induced arthritis, a decrease in CRH synthesis and secretion
has also been observed, together with a very marked increase The Importance of Pulsatility for
in pituitary portal levels of vasopressin (2,34,68). The lack of Hormonal and Behavioral Response
changes in corticosterone pulsatility across the 24-h cycle in to Stress
these animals suggests a consistent level of CRH (and AVP)
activation of the pituitary across the day within the range that The functional interaction between glucocorticoid pulsatility
will generate pulsatile ACTH and corticosterone secretion, and the response to stress has been investigated in a number of
as predicted by the mathematical model described by Walker studies where rats are exposed to stress during different phases
and colleagues (201, 202). of their ultradian corticosterone secretory profile (165, 213).
Another model of chronic stress that has been found to Windle and colleagues (213) have shown that the timing of
affect corticosterone pulsatility in the rat is exposure to con- the stressor relative to the phase of the underlying ultradian
stant light. Experimental animals are normally kept under a rhythm is crucial in determining the magnitude of the cor-
12-h light-dark cycle, with light input acting as a zeitgeber ticosterone response. Specifically, in rats exposed to a mild
processed through the SCN. Disruption of the SCN signal, stressor such as white noise, the corticosterone response is
either by physical lesioning of the SCN or by disruption of considerably greater when the stressor is applied during the
the light input, induces a loss of circadian rhythmicity and rising phase of a corticosterone pulse than when the stressor
this is associated with changes in corticosterone pulsatility. is applied during the falling phase (Fig. 10), which suggests
Rats maintained under conditions of constant light for a pro- a facilitated stress response during the rising phase and/or
longed period of time (5 weeks) lose their circadian corticos- an inhibitory effect during the falling phase. This relation-
terone rhythm, which is due to increased pulsatility during the ship between pulse-phase and the ability of the HPA axis
nadir phase of the circadian cycle, with no difference between to respond to acute stress has been observed in both male
the nadir and the peak phase pulse amplitude (200). These and female rats. These observations indicate the existence of
same rats have increased levels of CRH mRNA in the morn- a dynamic interaction between basal glucocorticoid pulsatil-
ing, which probably accounts for the increased corticosterone ity and the ability of an animal to mount a hormonal stress
secretion during the nadir phase. This suggests that removal response.
of the inhibitory SCN input to the hypothalamic PVN results Mathematical modeling and in vivo experimental work
in sufficient CRH secretion throughout the 24 h to maintain suggest that ultradian glucocorticoid oscillations can be gen-
ultradian pituitary-adrenal activity. erated by the feedforward-feedback interaction between the
The SCN also mediates corticosterone secretion by regu- anterior pituitary and adrenal cortex, independent of pulsatile
lating adrenal sensitivity to ACTH through a mechanism that hypothalamic stimulation (201, 202). Therefore, exposure to
involves the ANS and splanchnic nerve. It has been shown a stressor, and its associated release of hypothalamic CRH
that light can activate adrenal secretion of corticosterone in and AVP, can be considered as a transient perturbation to the
a way that is independent of pituitary release of ACTH, but endogenous oscillatory activity of the pituitary-adrenal sys-
that involves activation of the splanchnic nerve (19, 81). It is tem. This mathematical model has also been used to under-
therefore possible that the effect of constant light exposure on stand in more detail the interaction between pulsatility and
corticosterone pulsatility may also occur due to a direct effect stress responsiveness (154). In addition to explaining ear-
on the adrenal gland. lier observations that the magnitude of the stress response
depends on the timing of the stress, analysis of this model
has also shown that an external stress can act as a resetting
Pathological changes: Neonatal programming mechanism to the phase of the endogenous ultradian rhythm.
Exposure to stress during the neonatal period has profound The mechanism underlying the differential HPA axis
effects on physiological functions that can be observed in response to stress in relationship to the ultradian corticos-
adult life. This phenomenon is known as neonatal program- terone rhythm has been further investigated in adrenalec-
ming. Early life stress can also program the development of tomized rats in which the endogenous hormone is replaced
the HPA axis with changes that persist for the rest of the life with an IV infusion of hourly pulses of corticosterone (165).

1286 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

(A)
600
Differential response to stress is associated with
changes in the ultradian glucocorticoid rhythm
Corticosterone (% of basal)

Both experimental data and mathematical modeling have


shown that a facilitated corticosterone response to stress
300
occurs when the stressor is applied during the rising phase of
an ultradian pulse (i.e., when the HPA axis is already secret-
ing hormone), or during an interpulse period (when the axis
is quiescent). In contrast, the axis is inhibited and unable to
respond when the stress is applied during the falling phase of
0
a pulse (154, 214). It is clear therefore that the characteristics
–60 –40 –20 0 20 40 60 of the corticosterone ultradian rhythm are important not only
Time (min)
for basal corticosterone secretion but also for maintaining
(B) hormonal responsiveness to stress. In light of this, the rela-
600
tionship between the timing of the exposure to stress relative
Corticosterone (% of basal)

to the phase of the corticosterone pulse has been investigated


in a number of experimental models where differences in cor-
ticosterone pulsatility have been observed, such as in rats of
300
different strain (213), reproductive stage (173, 211), gender
(169-172), and in rats exposed to chronic stress (174, 212).
Differences in the pattern of ultradian rhythmicity have
been observed between female Fisher and Lewis rats, and this
0 is reflected in differences in the corticosterone response to
–60 –40 –20 0 20 40 60 stress in these animals (213). The corticosterone response to
Time (min) noise stress is greater in the Fischer than in the Lewis female
rat. In the female Lewis rat, stress-induced corticosterone
Figure 10 Stress responsiveness is dependent on the phase of
the ultradian corticosterone rhythm. Corticosterone response in rats secretion does not occur when the noise is applied during the
exposed to a noise stress (10 min, 114 dB); blood samples were col- falling phase of the ultradian pulse. In contrast, female Fisher
lected every 10 min using an automated blood sampling system. Rats rats respond equally to a noise stress regardless of when it
stressed during the rising phase of an endogenous corticosterone pulse
(A) show much greater corticosterone responses than animals stressed occurs in relation to the phase of the endogenous corticos-
during the falling phase (B). Reproduced with permission from (214). terone rhythm. A pronounced difference in the duration of
stress-induced corticosterone release has also been observed
between the two strains, with Fischer rats showing a more
prolonged response.
Differential responses to stress have been observed fol-
Consistent with Windle et al. (214), exposure of these rats to lowing manipulation of the gonadal steroid environment of
noise stress results in an increase in ACTH, and this response both male and female rats, and changes in the ultradian rhythm
is more pronounced when the stressor is applied during the are associated with an altered response to both psychological
rising phase of the corticosterone ultradian pulse than during noise stress and LPS-induced immune challenge (169-172).
the falling phase. The differential ACTH response to noise Although there is a significant and prolonged increase in
stress, relative to the phase of corticosterone pulse, is also plasma corticosterone in both males and females in response
associated with a different behavioral response to the stressor. to noise and LPS, the amplitude of the response is higher
Indeed, the behavioral response of rats exposed to noise stress in females compared to males. Furthermore, gonadectomy
is higher in rats that are stressed during the rising phase of reverses this pattern with corticosterone profiles and stress-
the corticosterone pulse, compared to rats exposed to stress induced peak corticosterone release in castrated males that are
during the falling phase of the pulse. Interestingly, in addi- similar to those seen in sham females. In contrast, the effect of
tion to a differential hormonal and behavioral response to ovariectomy results in an ultradian profile and stress-induced
noise relative to the phase of the corticosterone pulse, differ- peak response similar to those in sham males (169). As seen
ential neuronal activation in the amygdala occurs when the for basal corticosterone pulsatility, the effects of gonadectomy
noise stress is delivered during the rising phase rather than on the stress response in both male and female are reversed
the falling phase of the ultradian pulse (165). These findings by androgen and oestradiol replacement in male and female,
are consistent with earlier behavioral studies showing that respectively (170). Similarly, neonatal masculinization, which
rats exposed to a male intruder during the rising phase of an results in reduced pulse amplitude, number, and frequency in
endogenous corticosterone pulse are more aggressive than rats adult female rats, is associated with reduced responsiveness to
exposed to the intruder during the falling phase of the pulse both noise and LPS stress, as seen in normal adult males (171).
(65, 66). In contrast, pre and postnatal deprivation of testosterone in

Volume 4, July 2014 1287


HPA Axis-Rhythms Comprehensive Physiology

male rats, which results in increased pulse number, amplitude unbound and free to diffuse across cell membranes and bind to
and frequency, is associated with significantly elevated cor- intracellular GRs and MRs in target tissues. Therefore, steroid
ticosterone secretion after noise stress or LPS administration hormones bound to carrier proteins are considered biologi-
(172). cally inactive and provide a reservoir of inactive circulating
Differential responses to stress have also been observed in hormone, and the levels of CBG regulate the amount of free
female rats at different reproductive stages (173, 211). While hormone available for diffusion into tissue. CBG is secreted
in virgin rats noise stress causes a rapid increase in plasma from hepatocytes (103) with a circadian pattern in the plasma
corticosterone concentration, in the lactating group, which that is in antiphase with the circadian variation in plasma cor-
has an increased number of pulses across the 24-h cycle, ticosterone levels (77, 114). This results in an increased level
no effects on plasma corticosterone levels are seen follow- of free corticosterone during the circadian peak. In addition,
ing noise stress. Furthermore, in the experimentally weaned CBG is also released from the liver in response to acute stress
group, which show lower basal corticosterone levels and a in parallel with the adrenal release of corticosterone, and this
lower number of pulses, the response to noise is of similar is thought to be important for maintaining low levels of free
amplitude, but more prolonged, compared to the virgin group corticosterone at target tissues (53, 152).
(211). With respect to the role of CBG in regulating the ultradian
A different response to stress has been observed in rats rhythm of active free unbound glucocorticoid, CBG acts to
exposed to chronic inflammation (adjuvant-induced rheuma- accentuate pulse profiles at the tissue level. In fact, it is known
toid arthritis). In these rats, the relationship between the pulse that in human, CBG has a relatively low saturation point: 400
phase and the timing of the stress is maintained through- to 500 nmol/L cortisol (12). Therefore, when levels of hor-
out the development of the inflammation (212). However, the mone are high, such as during the peak of each ultradian
overall corticosterone response to noise stress is significantly pulse, CBG is already saturated. This results in pulses of
lower in symptomatic rats than in controls. Thus, it is possi- free cortisol that are superimposed on pulses of total corti-
ble that as the number of pulses increases and the interpulse sol in which only about 10% is in the free state. This type
periods reduce in symptomatic rats, there is a greater propor- of augmented ultradian rhythm of corticosterone has been
tion of time when the rats are unable to respond to stress. demonstrated experimentally in the rat (53).
These data provide evidence for a direct relationship between It has also been shown in the rat that free corticosterone
increased basal HPA axis activity, associated with chronic levels in the blood show distinct circadian and ultradian
disease, and a decreased response to acute stress, and this rhythms, with a pulse frequency of approximately one pulse
is indeed consistent with previous data showing a decreased per hour (151), consistent with the pulse characteristics of
corticosterone response to acute stress in rats with adjuvant- total glucocorticoid hormone in plasma (214). Given that the
induced arthritis (2, 69), and also in humans with rheuma- level of free glucocorticoid depends on the availability of CBG
toid arthritis (33). Interestingly, there is no difference in the and its affinity for the hormone, a recent study has investigated
corticosterone response to LPS between rats with adjuvant- whether there are dynamic changes in the affinity of cortisol
induced arthritis and control rats. for CBG in human (23). This study has shown that the affinity
Changes in basal corticosterone pulsatility in rats neona- of CBG for cortisol is remarkably temperature sensitive—
tally exposed to an inflammatory stress are also associated the affinity of cortisol for CBG drops approximately 16-fold
with a differential corticosterone response to stress (174). as the temperature increases from 35 to 42◦ C. Interestingly,
Rats that receive endotoxin in early life show an increase in there are circadian and ultradian differences in core temper-
both the amplitude and number of corticosterone pulses dur- ature that can be between 1 and 4◦ C in rodents and around
ing basal conditions in adult life, and are also hyper-reactive 1◦ C in man. Based on the findings of Cameron et al. (23),
to stress (noise and LPS). these changes in temperature may be sufficient to cause rapid
changes in the availability of free glucocorticoid over the
course of both the circadian and ultradian rhythms of hormone
Ultradian Rhythm of Glucocorticoids secretion.
in Target Tissues Temperature dependence of glucocorticoids binding to
CBG is also an important factor for the free/bound hormone
CBG regulates the level of glucocorticoids at ratio in situations when robust changes in body temperature
target tissues occur. For example, an increase in body temperature of as little
Once they have been released into the general circulation, as 1◦ C in febrile patients may have profound effects, markedly
approximately 95% of glucocorticoid molecules become increasing the amplitude of free pulsatile bio-available gluco-
bound to carrier proteins, which prevents the steroid from corticoid at the tissue and cellular level. Indeed, studies in rats
diffusing into cells in target tissues. In the rat, approximately have shown changes in the ultradian rhythm of corticosterone
80% of circulating corticosterone is bound to corticosteroid- secretion in animals exposed to inflammation (174,212), sug-
binding globulin (CBG), and 10% to 15% is bound to albumin gesting that changes in the body temperature of rats exposed to
(113). The remaining fraction of circulating corticosterone is inflammatory stress may have an effect on the corticosterone

1288 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

0.4

Hippocampal free corticosterone (μg/dL)


0.3

0.2

0.1

0
0700 1300 1900 0100 0700
Clock time (h)

Figure 11 Ultradian rhythm of hippocampal free corticosterone measured by in vivo micro-


dialysis in a freely behaving male Wistar rat. Hippocampal dialysate samples were collected every
10 min between 0800 and 2200 h and every 30 min between 2200 and 0800 h. Male Wistar rats
show a clear and distinct circadian and ultradian pattern of corticosterone in the hippocampus
with a pulse frequency that is similar to that observed in plasma corticosterone. Reproduced with
permission from (53).

binding to CBG, and thus on the pattern of free corticosterone most of the differences observed between male and female. In
pulsatility. addition to CBG, other factors contribute to the regulation of
corticosterone levels in the brain, including the ATP-binding
cassette transporter P-glycoprotein (Pgp) (99). However, to
Ultradian rhythm of glucocorticoid is date, there are no reported differences in Pgp levels between
maintained in target organs male and female rats.
Recent studies have shown that the ultradian rhythm of corti- The ultradian rhythm of free corticosterone is highly syn-
costerone secretion is not only observed in the blood but also chronized between the blood and the target tissues both in the
in tissue targets such as the brain and subcutaneous tissue rat and in man (14,152). Dual-probe microdialysis techniques
(52-54, 152). Using the technique of microdialysis in the rat, have been used to directly compare, within the same ani-
Droste et al. (53) have shown that the corticosterone ultradian mals, the corticosterone ultradian rhythm between the blood
rhythm can be detected in discrete brain regions such as the and the subcutaneous tissue, and between the blood and the
hippocampus and the striatum, mirroring the pulses detected hippocampus. These studies have shown that corticosterone
in the peripheral circulation. In the hippocampus, the fre- secretion is highly synchronized between the blood and both
quency of the pulses is approximately one per hour, which the subcutaneous tissue and the brain. However, although
fits well with the pulse frequency of corticosterone observed there are no differences in the pulse amplitude between the
in the blood circulation and in the adrenal gland (Fig. 11). blood, the subcutaneous tissue, and the brain during the nadir
Interestingly, in contrast with plasma corticosterone, the ultra- phase of the circadian cycle, free corticosterone levels are
dian rhythm of hippocampal corticosterone is not affected by slightly lower in target tissues, compared with the blood, dur-
gender (54). Indeed, while both plasma corticosterone levels ing the active phase.
and pulse amplitude are higher in female rats compared to As has been seen for plasma corticosterone, changes in the
male rats, there is no difference in either hippocampal cor- ultradian rhythm have also been observed in the hippocam-
ticosterone levels or pulse characteristics between male and pus. For example, there are changes in the ultradian and cir-
female. Although this discrepancy cannot be fully explained, cadian rhythms of free corticosterone concentrations in the
it should be noted that while the plasma corticosterone levels hippocampus of rats that have been exposed to long-term
reported by Windle and colleagues (214) and by Seale and col- (4 weeks) voluntary exercise (52). Specifically, exercising
leagues (169) represent both CBG-bound and free hormone, rats display increased pulse amplitudes and mean free corti-
the corticosterone levels in brain measured by Droste and costerone levels during the peak phase of the circadian cycle,
colleagues (53, 54) represent only the free biologically active compared with control animals during this period, but show
fraction. Therefore, given that the levels of plasma CBG are no differences in pulse frequency. In contrast, there are no dif-
higher (by up to 2.5-fold) in female than in male rats (60, ferences in ultradian rhythm characteristics between the two
125), the CBG-bound corticosterone fraction can account for groups during the nadir phase of the circadian cycle.

Volume 4, July 2014 1289


HPA Axis-Rhythms Comprehensive Physiology

In man, studies have also been performed showing ultra- The pattern of DNA binding has been investigated in more
dian and circadian rhythmicity in subcutaneous tissue (14). detail in the MCF7(3617) mouse cell line (181). This cell line
The same authors have now shown a close correlation between stably expresses GR protein fused to a green fluorescent pro-
total cortisol levels in the blood and free cortisol levels in the tein (GFP) tag, allowing visualization of receptor movement
blood and subcutaneous tissue (13). in the living cell. Additionally, these cells also contain an
expanded MMTV long terminal repeat giving an array of
approximately 1200 GRE sequences. Using live cell imaging
The Importance of Glucocorticoid and photobleaching techniques, Stavreva et al. have investi-
gated the real-time effect of pulses of corticosterone applied to
Pulsatility for GR Signaling these cells and visualized the intracellular effect of the treat-
Glucocorticoids act via two distinct intracellular receptors, ment on GR activity (Fig. 12A). They have found that each
MR and GR, which upon activation by ligands, translocate corticosterone pulse is paralleled by cyclic GFP-GR bind-
into the nucleus where they induce or repress gene transcrip- ing to the GREs and that dissociation of GFP-GR from GRE
tion. Given that GR is widely expressed throughout the body, occurs within 10 min from the beginning of the hormone
the effects of glucocorticoids at target organs are mediated washout period. Further studies in both AtT20 and MCF7
primarily by activation of GR. When glucocorticoid levels cells (40) have shown that this transient activation, typical
are low, GR is retained in the cytoplasm, where it is bound of normal ultradian rhythmicity, only occurs in response to
to chaperone molecules, including HSP90 and p23 (149). the endogenous glucocorticoids corticosterone and hydrocor-
These molecular chaperones hold GR in a conformation that tisone, whereas a single pulse of the synthetic glucocorticoid
exposes the ligand binding cleft, and therefore allow GR to
be activated by the ligand (150). Once activated, GR under-
goes conformational changes that induce dissociation from (A) Uninduced Pulse 1 Washout 1
the chaperone complex, leading to exposure of nuclear local-
ization sequences (NLS) in the GR hinge domain and ligand-
binding domain (140). These sequences are important so that
importin molecules can recognize and bind at these NLS
sequences, and facilitate active transport of GR through the
nuclear pore. Once in the nucleus, GR can dimerize and bind Pulse 2 Washout 2 Pulse 3
to specific regulatory sequences in the DNA, termed glu-
cocorticoid response elements (GREs) (168). This event is
followed by the recruitment of cofactors, including members
of the p160 family (129) and the histone acetyl transferase
(HAT) proteins P300 and CBP (105). Importantly, P300 and
CBP are involved in the reorganization of the local chro-
matin structure to allow access of RNA polymerase and the (B) 30
basal transcriptional machinery at the transcriptional start site Pulsatile
(203), ultimately leading to transcription of target genes. It is Constant
important to understand how the dynamic ultradian oscilla-
Fold change

20
tions in glucocorticoid levels we have described above affect
the activity of these receptors and their downstream influences
on gene transcription. 10

Effect of ultradian glucocorticoid rhythm on GR


0
activity: in vitro studies 0 20 40 60 80 100 120
The importance of glucocorticoid pulsatility for the physio- Time (min)
logical regulation of GR activity has been investigated in vitro
Figure 12 Ultradian corticosterone pulses induce pulsatile GR-GRE
in a number of cell lines, including mouse corticotroph AtT- interactions and pulsatile gene transcription. (A) Real time single cell
20, human HeLa, rat liver HTC, and mouse MCF7(3617) cell imaging of GFP-labeled GR loading at an engineered array of GREs
lines (38,40,181). Exposure of cells to pulses of ligand (corti- in the MCF-7 (3617) mouse cell line. Each pulse of corticosterone
causes a “wave” of GR-GRE binding (fluorescent green points) that is
sol for human HeLa cells and corticosterone for rat HTC and rapidly reversed (GR-GRE dissociation) following hormone withdrawal
mouse AtT-20 cells) results in cyclical GR activation. In all (washout). (B) Dependence of transcriptional dynamics on the pattern
three cell lines, each pulse of glucocorticoid initiates a “wave” of hormone stimulation. Ultradian corticosterone treatment results in
pulsatile transcription of the Period 1 gene (Per1) (black), whereas
of GR activation and DNA association. This GR-GRE asso- constant corticosterone induces sustained transcription (grey). Levels of
ciation decreases rapidly after hormone washout, returning to Per1 primary transcript were measured in the 3134 cell line by real-
baseline levels before addition of the next pulse 45 min later. time quantitative PCR. Data reproduced with permission from (181).

1290 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

analogue dexamethasone results in a prolonged GR activation cyclical transcriptional activity at the level of the Per1 pro-
profile. The dynamics of GR dissociation from, and reasso- moter involves cyclical intranuclear chaperone protein activ-
ciation with, the ligand, and the mechanism underlying these ity. Indeed, consistent with its effect on corticosterone binding
events, has also been investigated in MCF7 cells (181). When to GR, inhibition of HSP90 with geldanamycin results in com-
these cells are exposed to pulses of corticosterone, GR rapidly plete ablation of ultradian cyclical transcriptional activity at
dissociates from the ligand following washout. However, for the Per1 gene, as shown by lack of GR, CBP, or p300 recruit-
GR to be able to rebind the ligand when the next pulse of ment at the GRE and reduced acetylation and transcriptional
corticosterone arrives, and therefore initiate a second cycle rate (38).
of transcriptional activity, it requires the formation of a com-
plex with chaperone proteins in the nucleus. Indeed, when
the chaperone complex formation is blocked using the Hsp90 Effects of glucocorticoid pulsatility on
inhibitor geldanamycin, GR cannot bind corticosterone, and GR activity: in vivo studies
this results in GR failure to associate with GRE, and an accel- Given the importance of pulsatile glucocorticoid pulsatility
erated proteasome-mediated degradation of GR. for gene regulation in vitro, the question arises as to the
In other studies, it has been shown that pulsatile exposure importance of pulsatility for GR-mediated gene regulation
to corticosterone also results in dynamic recruitment of Poly- in the living animal. Therefore, the effect of pulsatile corti-
merase II (Pol II) at GRE sites, with periods of reduced Pol costerone in target tissue has been explored in detail using
II exchange with DNA that coincide with maximal GR-GRE adrenalectomized rats, in which corticosterone is replaced by
binding. Using chromatin immunoprecipitation (ChIP), it has hourly pulses delivered by IV injection (39, 41, 181).
been shown that in both MCF7 (181) and AtT20 (38) cells, The dynamics of GR and MR activation over each ultra-
consistent with the pattern of GR binding to DNA, there is dian pulse can be measured by determining nuclear translo-
also a pulsatile pattern of GR binding to GREs in the promoter cation of the receptors (39), and this has been achieved by
sequences of naturally occurring glucocorticoid-responsive using subcellular fractionation and Western blotting to detect
genes, including genes encoding metallothionein1 and Period GR and MR in cytoplasmic and nuclear compartments. Hip-
1 (Per1), and the negatively regulated pro-opiomelanocortin pocampal GR and MR have been found to be rapidly acti-
(38). This is also reflected in the pattern of transcription vated following each pulse. Following nuclear translocation
of a number of GR-responsive genes, as measured by real and binding to DNA, GR is rapidly cleared from the nucleus,
time quantitative PCR (RTqPCR) using primers designed to with nuclear levels dropping significantly within 30 min and
recognize nascent RNA transcripts (or heteronuclear RNA, returning to baseline levels by 60 min after each pulse. In
hnRNA). Pulsatile corticosterone exposure results in a rapid contrast, MR is not rapidly cleared from the nucleus, with
and transient increase in hnRNA from several GR-regulated levels remaining high for up to 60 min after each pulse. As
genes (38, 181), whereas constant exposure to corticosterone this timing is coincident with the duration of the physio-
is associated with a continuous increase in hnRNA (181) logical inter-pulse interval, these findings demonstrate that
(Fig. 12B). In addition to this, constant treatment with corti- in the hippocampus, pulsatile exposure to corticosterone is
costerone results in accumulation of higher levels of mature a sufficient stimulus to retain the high affinity MR in the
RNA (mRNA) and protein, when compared with pulsatile cor- nucleus, while causing the lower affinity GR to translocate
ticosterone (181). Interestingly, when the cells are treated with in and out of the nucleus, in parallel with fluctuating steroid
a synthetic glucocorticoid, such as Dexamethasone, which is levels.
known to have a higher affinity for GR—and therefore pro- The dynamics of nuclear localization of GR has been
longed GR association with GRE—the duration of the tran- investigated using immunofluorescent confocal imaging of
scriptional response is longer (181). GR in hippocampal CA1 neurons (Fig. 13A) (41). This study
The mechanism underlying the dynamics of pulsatile gene has shown that a rapid but transient increase in nuclear GR
transcription has been further investigated in AtT20 cells. In levels occurs in response to an IV corticosterone pulse. Prior
this case, GR-mediated cyclical transcription of the Per1 gene, to the pulse, GR immunoreactivity is predominantly cyto-
induced by a pulse of corticosterone, is associated with rapid, plasmic, whereas a significant nuclear translocation of GR is
but transient, generalized protein acetylation at the GRE site detectable at 15 min after the pulse. By 60 min after the pulse,
within the Per1 promoter, and ChIP assays have revealed the nuclear GR immunofluorescence has returned to baseline lev-
nucleosome core protein H4 as an acetylation target (38). els.
Furthermore, the HAT complex members CBP and p300 have The mechanism underlying the rapid nuclear clearance of
also been found to undergo cyclical recruitment to the GRE GR in the hippocampus, and therefore, its rapid inactivation
region in the Per1 promoter. The pattern of both CBP and mediated by proteosomal activity, has also been investigated
p300 recruitment at the Per1 promoter is consistent with the (39). Using the specific 26S proteasome inhibitor MG132,
pattern of both GR and RNA Pol II recruitment at the Per1 injected intra-cerebraventricularly, Conway-Campbell et al.
promoter region. In turn, the pattern of Pol II recruitment is have shown that irreversible proteasomal inhibition abolishes
consistent with the pattern of Per1 transcription in this cell the rapid clearance of GR from the nucleus, and thus restores
line (181). Furthermore, it has been shown that GR-mediated elevated levels of nuclear activated GR.

Volume 4, July 2014 1291


HPA Axis-Rhythms Comprehensive Physiology

(A)
0 min 15 min 60 min 120 min

(B) (C)

Per1 hnRNA (fold change)


800
Corticosterone (ng/mL)

2.0
600
1.5
400
1.0
200

0 0.5
0 1 60 61 120 121 180 181 0 30 60 90 120 150 180 210
Time (min) Time (min)

Figure 13 Corticosterone ultradian rhythm induces pulsatile GR activation and GR-mediated gene tran-
scription in the rat hippocampus. To determine the effect of pulsatile corticosterone treatment on GR nuclear
translocation and transcription of the Period 1 (Per1) gene, multiple corticosterone pulses (100ng/pulse;
i.v.) were given at hourly intervals. (A) GR immunofluorescence in the CA1 region of the hippocampus
throughout the time course of one corticosterone pulse in adrenalectomized rats. At time 0 min, GR
immunoreactivity is predominantly cytoplasmic. A clear transient increase of approximately twofold can
be seen in nuclear GR within 15 min of the corticosterone injection, which returns to baseline levels by
60 min. (B) Circulating corticosterone levels induced by four corticosterone pulses in adrenalectomized
rats. Corticosterone is maximally increased in plasma at 1 min after each injection and then subsequently
cleared according to the characterized half-life of corticosterone in the blood. (C) Each corticosterone
pulse induces a pulse of Per1 gene transcription. Per1 primary transcript levels increase rapidly, reaching
a maximum at 30 min after each injection and, consistent with the pattern of GR dissociation from the
DNA as corticosterone is cleared from the circulation, return to basal levels at 60 min after each pulse.
Reproduced with permission from (41).

Consistent with the effect of pulses of corticosterone on intrinsically dependent on the secretion pattern of corticos-
cyclic GR nuclear translocation, pulsatile corticosterone infu- terone released from the adrenal gland. This is very important
sions induce “waves” of GR activation and DNA binding in given that glucocorticoids in the liver regulate numerous vital
the hippocampus of rats (41). Using ChIP, this has been con- functions, including glucose metabolism.
firmed by showing that the pulses of GR activation result in
cyclical recruitment of GR to a GRE in the promoter of the
Per1 gene. Furthermore, each pulse of corticosterone induces Nongenomic effects of glucocorticoid pulsatility
a pulse of transcription of Per1, as shown by measurements of A number of studies have shown that the ultradian rhythm
Per1 hnRNA (Fig 13B). Interestingly, despite a pulsatile pat- of glucocorticoids is important for the transcription of GR-
tern of hnRNA, pulsatile corticosterone administration results regulated genes in glucocorticoid responsive target organs.
in an increase in Per1 mRNA that reaches constant levels, sug- In addition to their genomic effects, glucocorticoids also
gesting that ultradian activity with pulses at hourly intervals have rapid nongenomic effects on target tissues, such as
is optimized to maintain Per1 protein level at a steady state. rapid effects on neuronal activity in the brain (57, 74, 193).
In addition to the hippocampus, the effect of glucocor- Indeed, rapid effects of corticosterone on excitatory and
ticoid pulsatility on GR-induced gene transcription has also inhibitory inputs to the hippocampus have been described
been investigated in the liver (181). In a similar manner, pul- (101). Recently, rapid nongenomic effects of glucocorticoid
satile corticosterone induces cyclic GR-GRE binding, and pulsatility on neuronal activity have been studied using elec-
this is associated with pulsatile binding of GR to regulatory trophysiology techniques (100). Using a paired-pulse stimula-
regions in the target clock gene Period1 (Per1). This in turn tion paradigm, Karst and colleagues have shown that exposure
is followed by cyclic changes in Per1 hnRNA levels over the to two pulses of corticosterone increases mEPSC frequency
duration of the corticosterone pulse. As seen for GR dynamic in CA1 hippocampal pyramidal cells, and that this effect is
activity in the hippocampus, these results indicate that in comparable for both pulses. In contrast, when two corticos-
the liver, glucocorticoid signaling is highly dynamic and terone pulses are applied in the basolateral amygdala nucleus,

1292 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

neurons in this region respond to the first pulse with increased oscillating pituitary-adrenal network. On a slower time scale,
mEPSC frequency—but display a reduced mEPSC frequency circadian rhythmicity entrained by the SCN modifies these
in response to the second pulse (100). rapidly oscillating levels of glucocorticoid to create a pattern
of pulses that vary in amplitude throughout the day. These
pulsatile patterns of hormone are vital for maintaining home-
ostasis, stress responsiveness, and optimal metabolic and cog-
Effect of Disrupted Ultradian Rhythm nitive function.
on Hormonal and Behavioral
Response to Stress
The existence of an interaction between hormonal and behav- Acknowledgements
ioral responsiveness to stress and the ultradian rhythm of cor-
ticosterone has been shown in the rat (65, 66, 165, 214). The The work from Stafford Lightman and colleagues described
mechanism underlying the differential HPA axis response to in this review has been supported by grants from the Welcome
stress in relationship to the ultradian corticosterone rhythm Trust, the Medical Research Council (MRC), the Biotechnol-
has been further investigated in adrenalectomized rats in ogy and Biological Sciences Research Council (BBSRC), the
which corticosterone is replaced either as a pulsatile or con- Neuroendocrinology Charitable Trust, and the Engineering
stant infusion (165). In rats infused with constant levels of and Physical Sciences Research Council (EPSRC).
corticosterone, the ACTH response to stress is suppressed,
when compared to rats infused with either vehicle or pul-
satile corticosterone. Consistent with these data, disruption References
of the ultradian pattern of corticosterone secretion results in
changes in GR receptor sensitivity with profound effects on 1. Abe K, Kroning J, Greer MA, Critchlow V. Effects of destruction of
the suprachiasmatic nuclei on the circadian rhythms in plasma corti-
the glucocorticoid responsiveness of target tissues (166). In costerone, body temperature, feeding and plasma thyrotropin. Neuroen-
rats implanted with a corticosterone pellet (to abolish corti- docrinology 29: 119-131, 1979.
2. Aguilera G, Jessop DS, Harbuz MS, Kiss A, Lightman SL. Differential
costerone pulsatility), GR in the hippocampus is downregu- regulation of hypothalamic pituitary corticotropin releasing hormone
lated. This downregulation results in decreased GR nuclear receptors during development of adjuvant-induced arthritis in the rat.
J Endocrinol 153: 185-191, 1997.
translocation and transcription of the GR target gene GILZ 3. Ahima RS, Harlan RE. Charting of type II glucocorticoid receptor-like
in hippocampal CA1 cells in response to a corticosterone immunoreactivity in the rat central nervous system. Neuroscience 39:
579-604, 1990.
injection. 4. Akana SF, Cascio CS, Du JZ, Levin N, Dallman MF. Reset of feed-
back in the adrenocortical system: An apparent shift in sensitivity of
adrenocorticotropin to inhibition by corticosterone between morning
and evening. Endocrinology 119: 2325-2332, 1986.
Clinical relevance 5. Alexander LD, Sander LD. Vasoactive intestinal peptide stimulates
ACTH and corticosterone release after injection into the PVN. Regul
It is clear that for optimal transcriptional and nongenomic Pept 51: 221-227, 1994.
responses tissues need to be exposed to oscillating concentra- 6. Antoni FA. Hypothalamic control of adrenocorticotropin secretion:
Advances since the discovery of 41-residue corticotropin-releasing fac-
tions of cortisol. Current hormone replacement regimes fail tor. Endoc Rev 7: 351-378, 1986.
in two respects. First, they do not provide an anticipatory 7. Antoni FA, Holmes MC, Makara GB, Karteszi M, Laszlo FA. Evi-
dence that the effects of arginine-8-vasopressin (AVP) on pituitary cor-
burst of cortisol release prior to awakening, which is impor- ticotropin (ACTH) release are mediated by a novel type of receptor.
tant to ensure that cognitive and metabolic functions are ready Peptides 5: 519-522, 1984.
8. Apostolakis EM, Longo LD, Veldhuis JD, Yellon SM. Dissociation of
for the activities needed first thing in the morning. Second, pulsatile cortisol and adrenocorticotropin secretion in fetal sheep during
oral replacement therapy results in smooth levels of steroid late gestation. Endocrinology 130: 2571-2578, 1992.
9. Arakane F, King SR, Du Y, Kallen CB, Walsh LP, Watari H, Stocco
throughout the day, which do not provide ultradian regulation DM, Strauss JF, II. Phosphorylation of steroidogenic acute regulatory
for GR signaling mechanisms. It is perhaps not surprising protein (StAR) modulates its steroidogenic activity. J Biol Chem 272:
32656-32662, 1997.
that patients on glucocorticoid replacement have double the 10. Arriza JL, Simerly RB, Swanson LW, Evans RM. The neuronal miner-
age related mortality and, often, severe morbidity in terms alocorticoid receptor as a mediator of glucocorticoid response. Neuron
1: 887-900, 1988.
of lassitude, tiredness and poor cognitive function. Similarly, 11. Artemenko IP, Zhao D, Hales DB, Hales KH, Jefcoate CR. Mitochon-
little appreciation has gone into the timing of glucocorticoid drial processing of newly synthesized steroidogenic acute regulatory
protein (StAR), but not total StAR, mediates cholesterol transfer to
therapy for inflammatory or malignant disease to maximize cytochrome P450 side chain cleavage enzyme in adrenal cells. J Biol
effect and minimize side effects. Chem 276: 46583-46596, 2001.
12. Ballard PL. Delivery and transport of glucocorticoids to target cells.
Monogr Endocrinol 12: 25-48, 1979.
13. Bhake R LJ, Linthorst ACE, Lightman SL. Continuous ambulatory
free cortisol profiles. In: Abstract of the 43rd Annual Meeting of the
Conclusions International Society of Psychoneuroendocrinology (ISPNE). Leiden,
The Netherlands.: 2013.
14. Bhake RC, Leendertz JA, Linthorst AC, Lightman SL. Automated 24-
The HPA axis is a highly dynamic system. Ultradian rhyth- hours sampling of subcutaneous tissue free cortisol in humans. J Med
Eng Technol 37: 180-184, 2013.
micity emerges due to the interaction between the dynamic 15. Bingaman EW, Magnuson DJ, Gray TS, Handa RJ. Androgen inhibits
release of hypothalamic hormones and an intrinsically the increases in hypothalamic corticotropin-releasing hormone (CRH)

Volume 4, July 2014 1293


HPA Axis-Rhythms Comprehensive Physiology

and CRH-immunoreactivity following gonadectomy. Neuroendocrinol- 40. Conway-Campbell BL, Pooley JR, Hager GL, Lightman SL. Molec-
ogy 59: 228-234, 1994. ular dynamics of ultradian glucocorticoid receptor action. Mol Cell
16. Bittman EL, Doherty L, Huang L, Paroskie A. Period gene expression Endocrinol 348: 383-393, 2012.
in mouse endocrine tissues. Am J Physiol Regul Integr Comp Physiol 41. Conway-Campbell BL, Sarabdjitsingh RA, McKenna MA, Pooley JR,
285: R561-R569, 2003. Kershaw YM, Meijer OC, De Kloet ER, Lightman SL. Glucocorticoid
17. Buijs RM, Kalsbeek A, van der Woude TP, van Heerikhuize JJ, Shinn ultradian rhythmicity directs cyclical gene pulsing of the clock gene
S. Suprachiasmatic nucleus lesion increases corticosterone secretion. period 1 in rat hippocampus. J Neuroendocrinol 22: 1093-1100, 2010.
Am J Physiol 264: R1186-R1192, 1993. 42. Dallman MF, Akana SF, Cascio CS, Darlington DN, Jacobson L, Levin
18. Buijs RM, Van Eden CG. The integration of stress by the hypothala- N. Regulation of ACTH secretion: Variations on a theme of B. Recent
mus, amygdala and prefrontal cortex: Balance between the autonomic Prog Horm Res 43: 113-173, 1987.
nervous system and the neuroendocrine system. Prog Brain Res 126: 43. Dallman MF, Akana SF, Jacobson L, Levin N, Cascio CS, Shinsako J.
117-132, 2000. Characterization of corticosterone feedback regulation of ACTH secre-
19. Buijs RM, Wortel J, Van Heerikhuize JJ, Feenstra MG, Ter Horst GJ, tion. Ann N Y Acad Sci 512: 402-414, 1987.
Romijn HJ, Kalsbeek A. Anatomical and functional demonstration of 44. Dallman MF, Engeland WC, Rose JC, Wilkinson CW, Shinsako J,
a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway. Eur Siedenburg F. Nycthemeral rhythm in adrenal responsiveness to ACTH.
J Neurosci 11: 1535-1544, 1999. Am J Physiol 235: R210-R218, 1978.
20. Bunger MK, Wilsbacher LD, Moran SM, Clendenin C, Radcliffe LA, 45. Dallman MF, Jones MT. Corticosteroid feedback control of ACTH
Hogenesch JB, Simon MC, Takahashi JS, Bradfield CA. Mop3 is an secretion: Effect of stress-induced corticosterone ssecretion on subse-
essential component of the master circadian pacemaker in mammals. quent stress responses in the rat. Endocrinology 92: 1367-1375, 1973.
Cell 103: 1009-1017, 2000. 46. Dallmann R, Touma C, Palme R, Albrecht U, Steinlechner S. Impaired
21. Bush IE. Species differences in adrenocortical secretion. J Endocrinol daily glucocorticoid rhythm in Per1 (Brd) mice. J Comp Physiol [A]
9: 95-100, 1953. 192: 769-775, 2006.
22. Camacho-Arroyo I, Neri-Gomez T, Gonzalez-Arenas A, Guerra-Araiza 47. De Kloet ER. Hormones and the stressed brain. Ann N Y Acad Sci 1018:
C. Changes in the content of steroid receptor coactivator-1 and silencing 1-15, 2004.
mediator for retinoid and thyroid hormone receptors in the rat brain 48. de Kloet ER. Steroids, stability and stress. Front Neuroendocrinol 16:
during the estrous cycle. J Steroid Biochem Mol Biol 94: 267-272, 416-425, 1995.
2005. 49. de Kloet ER. Stress in the brain. Eur J Pharmacol 405: 187-198, 2000.
23. Cameron A, Henley D, Carrell R, Zhou A, Clarke A, Lightman S. 50. De Kloet ER, Vreugdenhil E, Oitzl MS, Joels M. Brain corticosteroid
Temperature-responsive release of cortisol from its binding globulin: A receptor balance in health and disease. Endoc Rev 19: 269-301, 1998.
protein thermocouple. J Clin Endocrinol Metab 95: 4689-4695, 2010. 51. Di S, Malcher-Lopes R, Halmos KC, Tasker JG. Nongenomic gluco-
24. Caraty A, Grino M, Locatelli A, Oliver C. Secretion of corticotropin corticoid inhibition via endocannabinoid release in the hypothalamus:
releasing factor (CRF) and vasopressin (AVP) into the hypophysial A fast feedback mechanism. J Neurosci 23: 4850-4857, 2003.
portal blood of conscious, unrestrained rams. Biochem Biophys Res 52. Droste SK, Collins A, Lightman SL, Linthorst AC, Reul JM. Distinct,
Commun 155: 841-849, 1988. time-dependent effects of voluntary exercise on circadian and ultradian
25. Carnes M, Brownfield MS, Kalin NH, Lent S, Barksdale CM. Episodic rhythms and stress responses of free corticosterone in the rat hippocam-
secretion of ACTH in rats. Peptides 7: 219-223, 1986. pus. Endocrinology 150: 4170-4179, 2009.
26. Carnes M, Kalin NH, Lent SJ, Barksdale CM, Brownfield MS. Pulsatile 53. Droste SK, de Groote L, Atkinson HC, Lightman SL, Reul JM, Linthorst
ACTH secretion: Variation with time of day and relationship to cortisol. AC. Corticosterone levels in the brain show a distinct ultradian rhythm
Peptides 9: 325-331, 1988. but a delayed response to forced swim stress. Endocrinology 149: 3244-
27. Carnes M, Lent S, Feyzi J, Hazel D. Plasma adrenocorticotropic hor- 3253, 2008.
mone in the rat demonstrates three different rhythms within 24 h. Neu- 54. Droste SK, de Groote L, Lightman SL, Reul JM, Linthorst AC. The
roendocrinology 50: 17-25, 1989. ultradian and circadian rhythms of free corticosterone in the brain are
28. Carnes M, Lent SJ, Erisman S, Barksdale C, Feyzi J. Immunoneutral- not affected by gender: An in vivo microdialysis study in Wistar rats.
ization of corticotropin-releasing hormone prevents the diurnal surge J Neuroendocrinol 21: 132-140, 2009.
of ACTH. Life Sci 45: 1049-1056, 1989. 55. Engler D, Pham T, Fullerton MJ, Clarke IJ, Funder JW. Evidence for an
29. Carnes M, Lent SJ, Erisman S, Feyzi J. Changes in mean plasma ACTH ultradian secretion of adrenocorticotropin, beta-endorphin and alpha-
reflect changes in amplitude and frequency of secretory pulses. Life Sci melanocyte-stimulating hormone by the ovine anterior and intermediate
43: 1785-1790, 1988. pituitary. Neuroendocrinology 49: 349-360, 1989.
30. Chalmers DT, Lovenberg TW, De Souza EB. Localization of novel 56. Engler D, Pham T, Liu JP, Fullerton MJ, Clarke IJ, Funder JW. Studies
corticotropin-releasing factor receptor (CRF2) mRNA expression to of the regulation of the hypothalamic-pituitary-adrenal axis in sheep
specific subcortical nuclei in rat brain: Comparison with CRF1 receptor with hypothalamic-pituitary disconnection. II. Evidence for in vivo
mRNA expression. J Neurosci 15: 6340-6350, 1995. ultradian hypersecretion of proopiomelanocortin peptides by the iso-
31. Chaudhary LR, Stocco DM. Effect of different steroidogenic stimuli on lated anterior and intermediate pituitary. Endocrinology 127: 1956-
protein phosphorylation and steroidogenesis in MA-10 mouse Leydig 1966, 1990.
tumor cells. Biochim Biophys Acta 1094: 175-184, 1991. 57. Evanson NK, Tasker JG, Hill MN, Hillard CJ, Herman JP. Fast feed-
32. Cherrington AD. Banting Lecture 1997. Control of glucose uptake and back inhibition of the HPA axis by glucocorticoids is mediated by
release by the liver in vivo. Diabetes 48: 1198-1214, 1999. endocannabinoid signaling. Endocrinology 151: 4811-4819, 2010.
33. Chikanza IC, Chrousos G, Panayi GS. Abnormal neuroendocrine 58. Fahrenkrug J, Hannibal J, Georg B. Diurnal rhythmicity of the canonical
immune communications in patients with rheumatoid arthritis. Eur clock genes Per1, Per2 and Bmal1 in the rat adrenal gland is unaltered
J Clin Invest 22: 635-637, 1992. after hypophysectomy. J Neuroendocrinol 20: 323-329, 2008.
34. Chowdrey HS, Larsen PJ, Harbuz MS, Jessop DS, Aguilera G, Eckland 59. Fulkerson WJ. Synchronous episodic release of cortisol in the sheep.
DJ, Lightman SL. Evidence for arginine vasopressin as the primary J Endocrinol 79: 131-132, 1978.
activator of the HPA axis during adjuvant-induced arthritis. Br J Phar- 60. Gala RR, Westphal U. Corticosteroid-binding globulin in the rat: Stud-
macol 116: 2417-2424, 1995. ies on the sex difference. Endocrinology 77: 841-851, 1965.
35. Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune- 61. Gekakis N, Staknis D, Nguyen HB, Davis FC, Wilsbacher LD, King DP,
mediated inflammation. N Engl J Med 332: 1351-1362, 1995. Takahashi JS, Weitz CJ. Role of the CLOCK protein in the mammalian
36. Churchill PF, Kimura T. Topological studies of cytochromes P-450scc circadian mechanism. Science 280: 1564-1569, 1998.
and P-45011 beta in bovine adrenocortical inner mitochondrial mem- 62. Gillies GE, Linton EA, Lowry PJ. Corticotropin releasing activity of
branes. Effects of controlled tryptic digestion. J Biol Chem 254: 10443- the new CRF is potentiated several times by vasopressin. Nature 299:
10448, 1979. 355-357, 1982.
37. Clark RG, Chambers G, Lewin J, Robinson IC. Automated repetitive 63. Girotti M, Pace TW, Gaylord RI, Rubin BA, Herman JP, Spencer RL.
microsampling of blood: Growth hormone profiles in conscious male Habituation to repeated restraint stress is associated with lack of stress-
rats. J Endocrinol 111: 27-35, 1986. induced c-fos expression in primary sensory processing areas of the rat
38. Conway-Campbell BL, George CL, Pooley JR, Knight DM, Norman brain. Neuroscience 138: 1067-1081, 2006.
MR, Hager GL, Lightman SL. The HSP90 molecular chaperone cycle 64. Graham ES, Littlewood P, Turnbull Y, Mercer JG, Morgan PJ, Barrett
regulates cyclical transcriptional dynamics of the glucocorticoid recep- P. Neuromedin-U is regulated by the circadian clock in the SCN of the
tor and its coregulatory molecules CBP/p300 during ultradian ligand mouse. Eur J Neurosci 21: 814-819, 2005.
treatment. Mol Endocrinol 25: 944-954, 2011. 65. Haller J, Halasz J, Mikics E, Kruk MR, Makara GB. Ultradian corticos-
39. Conway-Campbell BL, McKenna MA, Wiles CC, Atkinson HC, de terone rhythm and the propensity to behave aggressively in male rats.
Kloet ER, Lightman SL. Proteasome-dependent down-regulation of J Neuroendocrinol 12: 937-940, 2000.
activated nuclear hippocampal glucocorticoid receptors determines 66. Haller J, Millar S, van de Schraaf J, de Kloet RE, Kruk MR. The active
dynamic responses to corticosterone. Endocrinology 148: 5470-5477, phase-related increase in corticosterone and aggression are linked.
2007. J Neuroendocrinol 12: 431-436, 2000.

1294 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

67. Handa RJ, Weiser MJ. Gonadal steroid hormones and the hypothalamo- 92. Jones MT, Tiptaft EM, Brush FR, Fergusson DA, Neame RL. Evi-
pituitary-adrenal axis. Front Neuroendocrinol 35: 197-220, dence for dual corticosteroid-receptor mechanisms in the feedback
2014. control of adrenocorticotrophin secretion. J Endocrinol 60: 223-233,
68. Harbuz MS, Rees RG, Eckland D, Jessop DS, Brewerton D, Lightman 1974.
SL. Paradoxical responses of hypothalamic corticotropin-releasing fac- 93. Kalsbeek A, Buijs RM, van Heerikhuize JJ, Arts M, van der Woude TP.
tor (CRF) messenger ribonucleic acid (mRNA) and CRF-41 peptide and Vasopressin-containing neurons of the suprachiasmatic nuclei inhibit
adenohypophysial proopiomelanocortin mRNA during chronic inflam- corticosterone release. Brain Res 580: 62-67, 1992.
matory stress. Endocrinology 130: 1394-1400, 1992. 94. Kalsbeek A, Fliers E, Hofman MA, Swaab DF, Buijs RM. Vasopressin
69. Harbuz MS, Rees RG, Lightman SL. HPA axis responses to acute and the output of the hypothalamic biological clock. J Neuroendocrinol
stress and adrenalectomy during adjuvant-induced arthritis in the rat. 22: 362-372, 2010.
Am J Physiol 264: R179-R185, 1993. 95. Kalsbeek A, van der Spek R, Lei J, Endert E, Buijs RM, Fliers E.
70. Hastings M, Maywood ES. Circadian clocks in the mammalian brain. Circadian rhythms in the hypothalamo-pituitary-adrenal (HPA) axis.
BioEssays 22: 23-31, 2000. Mol Cell Endocrinol 349: 20-29, 2012.
71. Hastings M, O’Neill JS, Maywood ES. Circadian clocks: Regula- 96. Kalsbeek A, van der Vliet J, Buijs RM. Decrease of endogenous vaso-
tors of endocrine and metabolic rhythms. J Endocrinol 195: 187-198, pressin release necessary for expression of the circadian rise in plasma
2007. corticosterone: A reverse microdialysis study. J Neuroendocrinol 8:
72. Hendrickson AE, Wagoner N, Cowan WM. An autoradiographic and 299-307, 1996.
electron microscopic study of retino-hypothalamic connections. Z Zell- 97. Kalsbeek A, van Heerikhuize JJ, Wortel J, Buijs RM. A diurnal rhythm
forsch Mikrosk Anat 135: 1-26, 1972. of stimulatory input to the hypothalamo-pituitary-adrenal system as
73. Henley DE, Leendertz JA, Russell GM, Wood SA, Taheri S, Woltersdorf revealed by timed intrahypothalamic administration of the vasopressin
WW, Lightman SL. Development of an automated blood sampling V1 antagonist. J Neurosci 16: 5555-5565, 1996.
system for use in humans. J Med Eng Technol 33: 199-208, 2009. 98. Kaneko M, Hiroshige T, Shinsako J, Dallman MF. Diurnal changes
74. Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid- in amplification of hormone rhythms in the adrenocortical system. Am
mediated negative feedback, and regulation of the hypothalamic- J Physiol 239: R309-R316, 1980.
pituitary-adrenal axis. Neuroscience 204: 5-16, 2012. 99. Karssen AM, Meijer OC, van der Sandt IC, Lucassen PJ, de Lange
75. Hinz B, Hirschelmann R. Rapid non-genomic feedback effects of glu- EC, de Boer AG, de Kloet ER. Multidrug resistance P-glycoprotein
cocorticoids on CRF-induced ACTH secretion in rats. Pharm Res 17: hampers the access of cortisol but not of corticosterone to mouse and
1273-1277, 2000. human brain. Endocrinology 142: 2686-2694, 2001.
76. Holaday JW, Martinez HM, Natelson BH. Synchronized ultradian cor- 100. Karst H, Berger S, Erdmann G, Schutz G, Joels M. Metaplasticity of
tisol rhythms in monkeys: Persistence during corticotropin infusion. amygdalar responses to the stress hormone corticosterone. Proc Natl
Science 198: 56-58, 1977. Acad Sci U S A 107: 14449-14454, 2010.
77. Hsu BR, Kuhn RW. The role of the adrenal in generating the diurnal 101. Karst H, Berger S, Turiault M, Tronche F, Schutz G, Joels M. Miner-
variation in circulating levels of corticosteroid-binding globulin in the alocorticoid receptors are indispensable for nongenomic modulation of
rat. Endocrinology 122: 421-426, 1988. hippocampal glutamate transmission by corticosterone. Proc Natl Acad
78. Hubler TR, Scammell JG. Intronic hormone response elements mediate Sci U S A 102: 19204-19207, 2005.
regulation of FKBP5 by progestins and glucocorticoids. Cell Stress 102. Kasai M, Kannan H, Ueta Y, Osaka T, Inenaga K, Yamashita H. Effects
Chaperones 9: 243-252, 2004. of iontophoretically applied cortisol on tuberoinfundibular neurons in
79. Hugin-Flores ME, Steimer T, Aubert ML, Schulz P. Mineralo- and glu- hypothalamic paraventricular nucleus of anesthetized rats. Neurosci
cocorticoid receptor mrnas are differently regulated by corticosterone Lett 87: 35-40, 1988.
in the rat hippocampus and anterior pituitary. Neuroendocrinology 79: 103. Khan MS, Aden D, Rosner W. Human corticosteroid binding globulin
174-184, 2004. is secreted by a hepatoma-derived cell line. J Steroid Biochem 20:
80. Hum DW, Miller WL. Transcriptional regulation of human genes for 677-678, 1984.
steroidogenic enzymes. Clin Chem 39: 333-340, 1993. 104. King DP, Zhao Y, Sangoram AM, Wilsbacher LD, Tanaka M, Antoch
81. Ishida A, Mutoh T, Ueyama T, Bando H, Masubuchi S, Nakahara D, MP, Steeves TD, Vitaterna MH, Kornhauser JM, Lowrey PL, Turek
Tsujimoto G, Okamura H. Light activates the adrenal gland: Timing FW, Takahashi JS. Positional cloning of the mouse circadian clock
of gene expression and glucocorticoid release. Cell Metab 2: 297-307, gene. Cell 89: 641-653, 1997.
2005. 105. Kino T, Nordeen SK, Chrousos GP. Conditional modulation of gluco-
82. Iwasaki-Sekino A, Mano-Otagiri A, Ohata H, Yamauchi N, Shibasaki corticoid receptor activities by CREB-binding protein (CBP) and p300.
T. Gender differences in corticotropin and corticosterone secretion and J Steroid Biochem Mol Biol 70: 15-25, 1999.
corticotropin-releasing factor mRNA expression in the paraventricular 106. Kitchener P, Di Blasi F, Borrelli E, Piazza PV. Differences between
nucleus of the hypothalamus and the central nucleus of the amygdala in brain structures in nuclear translocation and DNA binding of the gluco-
response to footshock stress or psychological stress in rats. Psychoneu- corticoid receptor during stress and the circadian cycle. Eur J Neurosci
roendocrinology 34: 226-237, 2009. 19: 1837-1846, 2004.
83. Ixart G, Barbanel G, Nouguier-Soule J, Assenmacher I. A quantitative 107. Kow LM, Pfaff DW. Vasopressin excites ventromedial hypothala-
study of the pulsatile parameters of CRH-41 secretion in unanesthetized mic glucose-responsive neurons in vitro. Physiol Behav 37: 153-158,
free-moving rats. Exp Brain Res 87: 153-158, 1991. 1986.
84. Ixart G, Siaud P, Barbanel G, Mekaouche M, Givalois L, Assenmacher 108. Kruk MR, Halasz J, Meelis W, Haller J. Fast positive feedback between
I. Circadian variations in the amplitude of corticotropin-releasing hor- the adrenocortical stress response and a brain mechanism involved in
mone 41 (CRH41) episodic release measured in vivo in male rats: Corre- aggressive behavior. Behav Neurosci 118: 1062-1070, 2004.
lations with diurnal fluctuations in hypothalamic and median eminence 109. Lamia KA, Storch KF, Weitz CJ. Physiological significance of a periph-
CRH41 contents. J Biol Rhythms 8: 297-309, 1993. eral tissue circadian clock. Proc Natl Acad Sci U S A 105: 15172-15177,
85. Ixart G, Siaud P, Mekaouche M, Barbanel G, Givalois L, Assenmacher 2008.
I. Short-term but not long-term adrenalectomy modulates amplitude 110. Le Martelot G, Claudel T, Gatfield D, Schaad O, Kornmann B, Lo Sasso
and frequency of the CRH41 episodic release in push-pull cannulated G, Moschetta A, Schibler U. REV-ERBalpha participates in circadian
median eminence of free-moving rats. Brain Res 658: 185-191, 1994. SREBP signaling and bile acid homeostasis. PLoS Biol 7: e1000181,
86. Jasper MS, Engeland WC. Synchronous ultradian rhythms in adreno- 2009.
cortical secretion detected by microdialysis in awake rats. Am J Physiol 111. Leliavski A, Shostak A, Husse J, Oster H. Impaired glucocorticoid pro-
261: R1257-R1268, 1991. duction and response to stress in arntl-deficient male mice. Endocrinol-
87. Jasper MS, Engeland WC. Splanchnic neural activity modulates ultra- ogy 155: 133-142, 2014.
dian and circadian rhythms in adrenocortical secretion in awake rats. 112. Levi F, Schibler U. Circadian rhythms: Mechanisms and therapeutic
Neuroendocrinology 59: 97-109, 1994. implications. Annu Rev Pharmacol Toxicol 47: 593-628, 2007.
88. Jasper MS, Engeland WC. Splanchnicotomy increases adrenal sensi- 113. Lewis JG, Bagley CJ, Elder PA, Bachmann AW, Torpy DJ. Plasma free
tivity to ACTH in nonstressed rats. Am J Physiol 273: E363-E368, cortisol fraction reflects levels of functioning corticosteroid-binding
1997. globulin. Clin Chim Acta 359: 189-194, 2005.
89. Joels M, Urban IJ. The effect of microiontophoretically applied vaso- 114. Lewis JG, Mopert B, Shand BI, Doogue MP, Soule SG, Frampton CM,
pressin and oxytocin on single neurones in the septum and dorsal hip- Elder PA. Plasma variation of corticosteroid-binding globulin and sex
pocampus of the rat. Neurosci Lett 33: 79-84, 1982. hormone-binding globulin. Horm Metab Res 38: 241-245, 2006.
90. Jones MT, Brush FR, Neame RL. Characteristics of fast feedback con- 115. Li TZ, Wang CA, Chen YZ. [The rapid effect of the iontophoretically
trol of corticotrophin release by corticosteroids. J Endocrinol 55: 489- applied cortisol on unit activity of neurons in three brain areas in rats].
497, 1972. Sheng li xue bao : [Acta physiologica Sinica] 43: 280-285, 1991.
91. Jones MT, Hillhouse EW, Burden JL. Dynamics and mechanics of cor- 116. Lightman SL, Conway-Campbell BL. The crucial role of pulsatile activ-
ticosteroid feedback at the hypothalamus and anterior pituitary gland. ity of the HPA axis for continuous dynamic equilibration. Nat Rev
J Endocrinol 73: 405-417, 1977. Neurosci 11: 710-718, 2010.

Volume 4, July 2014 1295


HPA Axis-Rhythms Comprehensive Physiology

117. Lightman SL, Windle RJ, Julian MD, Harbuz MS, Shanks N, Wood 142. Orchinik M, Murray TF, Moore FL. A corticosteroid receptor in neu-
SA, Kershaw YM, Ingram CD. Significance of pulsatility in the HPA ronal membranes. Science 252: 1848-1851, 1991.
axis. Novartis Found Symp 227: 244-257; discussion 257-260, 2000. 143. Oster H, Damerow S, Hut RA, Eichele G. Transcriptional profiling in
118. Lin D, Sugawara T, Strauss JF, III, Clark BJ, Stocco DM, Saenger the adrenal gland reveals circadian regulation of hormone biosynthesis
P, Rogol A, Miller WL. Role of steroidogenic acute regulatory pro- genes and nucleosome assembly genes. J Biol Rhythms 21: 350-361,
tein in adrenal and gonadal steroidogenesis. Science 267: 1828-1831, 2006.
1995. 144. Oster H, Damerow S, Kiessling S, Jakubcakova V, Abraham D, Tian J,
119. Liu Y, Smith LI, Huang V, Poon V, Coello A, Olah M, Spiga F, Hoffmann MW, Eichele G. The circadian rhythm of glucocorticoids is
Lightman SL, Aguilera G. Transcriptional regulation of episodic glu- regulated by a gating mechanism residing in the adrenal cortical clock.
cocorticoid secretion. Mol Cell Endocrinol 371: 62-70, 2013. Cell Metab 4: 163-173, 2006.
120. Loh DH, Abad C, Colwell CS, Waschek JA. Vasoactive intestinal pep- 145. Park SY, Walker JJ, Johnson NW, Zhao Z, Lightman SL, Spiga F.
tide is critical for circadian regulation of glucocorticoids. Neuroen- Constant light disrupts the circadian rhythm of steroidogenic proteins
docrinology 88: 246-255, 2008. in the rat adrenal gland. Mol Cell Endocrinol 371: 114-123, 2013.
121. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid 146. Pfaff DW, Silva MT, Weiss JM. Telemetered recording of hormone
metabolism in humans: Fuelling fat redistribution in the metabolic effects on hippocampal neurons. Science 172: 394-395, 1971.
syndrome. J Endocrinol 197: 189-204, 2008. 147. Picard D. Intracellular dynamics of the Hsp90 co-chaperone p23 is
122. Mahmoud SN, Scaccianoce S, Scraggs PR, Nicholson SA, Gillham dictated by Hsp90. Exp Cell Res 312: 198-204, 2006.
B, Jones MT. Characteristics of corticosteroid inhibition of adreno- 148. Pon LA, Hartigan JA, Orme-Johnson NR. Acute ACTH regulation of
corticotrophin release from the anterior pituitary gland of the rat. adrenal corticosteroid biosynthesis. Rapid accumulation of a phospho-
J Endocrinol 102: 33-42, 1984. protein. J Biol Chem 261: 13309-13316, 1986.
123. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono 149. Pratt WB, Galigniana MD, Harrell JM, DeFranco DB. Role of hsp90
K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM. The and the hsp90-binding immunophilins in signalling protein movement.
nuclear receptor superfamily: The second decade. Cell 83: 835-839, Cell Signal 16: 857-872, 2004.
1995. 150. Pratt WB, Morishima Y, Osawa Y. The Hsp90 chaperone machinery
124. Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, regulates signaling by modulating ligand binding clefts. J Biol Chem
Ivanova G, Omura C, Mo S, Vitaterna MH, Lopez JP, Philipson LH, 283: 22885-22889, 2008.
Bradfield CA, Crosby SD, JeBailey L, Wang X, Takahashi JS, Bass 151. Qian X, Droste SK, Gutierrez-Mecinas M, Collins A, Kersante F, Reul
J. Disruption of the clock components CLOCK and BMAL1 leads to JM, Linthorst AC. A rapid release of corticosteroid-binding globulin
hypoinsulinaemia and diabetes. Nature 466: 627-631, 2010. from the liver restrains the glucocorticoid hormone response to acute
125. Mataradze GD, Kurabekova RM, Rozen VB. The role of sex steroids stress. Endocrinology 152: 3738-3748, 2011.
in the formation of sex-differentiated concentrations of corticosteroid- 152. Qian X, Droste SK, Lightman SL, Reul JM, Linthorst AC. Circadian
binding globulin in rats. J Endocrinol 132: 235-240, 1992. and ultradian rhythms of free glucocorticoid hormone are highly syn-
126. Maywood ES, O’Neill JS, Chesham JE, Hastings MH. Minireview: chronized between the blood, the subcutaneous tissue, and the brain.
The circadian clockwork of the suprachiasmatic nuclei–analysis of a Endocrinology 153: 4346-4353, 2012.
cellular oscillator that drives endocrine rhythms. Endocrinology 148: 153. Ralph MR, Foster RG, Davis FC, Menaker M. Transplanted suprachi-
5624-5634, 2007. asmatic nucleus determines circadian period. Science 247: 975-978,
127. McDearmon EL, Patel KN, Ko CH, Walisser JA, Schook AC, Chong 1990.
JL, Wilsbacher LD, Song EJ, Hong HK, Bradfield CA, Takahashi JS. 154. Rankin J, Walker JJ, Windle R, Lightman SL, Terry JR. Characteriz-
Dissecting the functions of the mammalian clock protein BMAL1 by ing dynamic interactions between ultradian glucocorticoid rhythmicity
tissue-specific rescue in mice. Science 314: 1304-1308, 2006. and acute stress using the phase response curve. PloS one 7: e30978,
128. McEwen BS. Physiology and neurobiology of stress and adaptation: 2012.
Central role of the brain. Physiol Rev 87: 873-904, 2007. 155. Reppert SM, Weaver DR. Coordination of circadian timing in mam-
129. Meijer OC, Steenbergen PJ, De Kloet ER. Differential expression and mals. Nature 418: 935-941, 2002.
regional distribution of steroid receptor coactivators SRC-1 and SRC-2 156. Reul JM, de Kloet ER. Two receptor systems for corticosterone in
in brain and pituitary. Endocrinology 141: 2192-2199, 2000. rat brain: Microdistribution and differential occupation. Endocrinology
130. Merriam GR, Wachter KW. Algorithms for the study of episodic hor- 117: 2505-2511, 1985.
mone secretion. Am J Physiol 243: E310-E318, 1982. 157. Reul JM, de Kloet ER. Anatomical resolution of two types of corti-
131. Mershon JL, Sehlhorst CS, Rebar RW, Liu JH. Evidence of a costerone receptor sites in rat brain with in vitro autoradiography and
corticotropin-releasing hormone pulse generator in the macaque computerized image analysis. J Steroid Biochem 24: 269-272, 1986.
hypothalamus. Endocrinology 130: 2991-2996, 1992. 158. Reul JM, de Kloet ER, van Sluijs FJ, Rijnberk A, Rothuizen J. Binding
132. Metherell LA, Chapple JP, Cooray S, David A, Becker C, Ruschendorf characteristics of mineralocorticoid and glucocorticoid receptors in dog
F, Naville D, Begeot M, Khoo B, Nurnberg P, Huebner A, Cheetham brain and pituitary. Endocrinology 127: 907-915, 1990.
ME, Clark AJ. Mutations in MRAP, encoding a new interacting partner 159. Rivier C, Vale W. Interaction of corticotropin-releasing factor and argi-
of the ACTH receptor, cause familial glucocorticoid deficiency type 2. nine vasopressin on adrenocorticotropin secretion in vivo. Endocrinol-
Nat Genet 37: 166-170, 2005. ogy 113: 939-942, 1983.
133. Mikami K, Strott CA. Cyclic AMP-dependent protein kinase activity 160. Rose JD, Moore FL, Orchinik M. Rapid neurophysiological effects
and protein phosphorylation in zones of the adrenal cortex. Biochem of corticosterone on medullary neurons: Relationship to stress-induced
Biophys Res Commun 138: 895-901, 1986. suppression of courtship clasping in an amphibian. Neuroendocrinology
134. Miller WL, Strauss JF, 3rd. Molecular pathology and mechanism of 57: 815-824, 1993.
action of the steroidogenic acute regulatory protein, StAR. J Steroid 161. Rotsztejn W, Lalonde J, Normand M, Fortier C. Feedback inhibi-
Biochem Mol Biol 69: 131-141, 1999. tion of adrenocorticotropin release by corticosterone infusions in the
135. Mohawk JA, Takahashi JS. Cell autonomy and synchrony of suprachi- adrenalectomized rat. Can J Physiol Pharmacol 53: 475-478, 1975.
asmatic nucleus circadian oscillators. Trends Neurosci 34: 349-358, 162. Rotsztejn WH, Normand M, Lalonde J, Fortier C. Relationship between
2011. ACTH release and corticosterone binding by the receptor sites of the
136. Moore RY, Lenn NJ. A retinohypothalamic projection in the rat. J Comp adenohypophysis and dorsal hippocampus following infusion of corti-
Neurol 146: 1-14, 1972. costerone at a constant rate in the adrenalectomized rat. Endocrinology
137. Mountjoy KG, Mortrud MT, Low MJ, Simerly RB, Cone RD. Local- 97: 223-230, 1975.
ization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and 163. Russell GM, Henley DE, Leendertz J, Douthwaite JA, Wood SA,
autonomic control circuits in the brain. Mol Endocrinol 8: 1298-1308, Stevens A, Woltersdorf WW, Peeters BW, Ruigt GS, White A, Veldhuis
1994. JD, Lightman SL. Rapid glucocorticoid receptor-mediated inhibition
138. Munck A, Guyre PM, Holbrook NJ. Physiological functions of gluco- of hypothalamic-pituitary-adrenal ultradian activity in healthy males.
corticoids in stress and their relation to pharmacological actions. Endoc J Neurosci 30: 6106-6115, 2010.
Rev 5: 25-44, 1984. 164. Saphier D, Feldman S. Iontophoretic application of glucocorticoids
139. Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U. Circadian inhibits identified neurones in the rat paraventricular nucleus. Brain
gene expression in individual fibroblasts: Cell-autonomous and self- Res 453: 183-190, 1988.
sustained oscillators pass time to daughter cells. Cell 119: 693-705, 165. Sarabdjitsingh RA, Conway-Campbell BL, Leggett JD, Waite EJ, Mei-
2004. jer OC, de Kloet ER, Lightman SL. Stress responsiveness varies over
140. Nishi M, Kawata M. Brain corticosteroid receptor dynamics and traf- the ultradian glucocorticoid cycle in a brain-region-specific manner.
ficking: Implications from live cell imaging. Neuroscientist 12: 119- Endocrinology 151: 5369-5379, 2010.
133, 2006. 166. Sarabdjitsingh RA, Isenia S, Polman A, Mijalkovic J, Lachize S, Dat-
141. Orchinik M, Murray TF, Franklin PH, Moore FL. Guanyl nucleotides son N, de Kloet ER, Meijer OC. Disrupted corticosterone pulsatile pat-
modulate binding to steroid receptors in neuronal membranes. Proc terns attenuate responsiveness to glucocorticoid signaling in rat brain.
Natl Acad Sci U S A 89: 3830-3834, 1992. Endocrinology 151: 1177-1186, 2010.

1296 Volume 4, July 2014


Comprehensive Physiology HPA Axis-Rhythms

167. Sawchenko PE, Swanson LW, Vale WW. Co-expression of 190. Takemori H, Kanematsu M, Kajimura J, Hatano O, Katoh Y, Lin XZ,
corticotropin-releasing factor and vasopressin immunoreactivity in par- Min L, Yamazaki T, Doi J, Okamoto M. Dephosphorylation of TORC
vocellular neurosecretory neurons of the adrenalectomized rat. Proc initiates expression of the StAR gene. Mol Cell Endocrinol 265-266:
Natl Acad Sci U S A 81: 1883-1887, 1984. 196-204, 2007.
168. Schoneveld OJ, Gaemers IC, Lamers WH. Mechanisms of glucocorti- 191. Takemori H, Okamoto M. Regulation of CREB-mediated gene expres-
coid signalling. Biochim Biophys Acta 1680: 114-128, 2004. sion by salt inducible kinase. J Steroid Biochem Mol Biol 108: 287-291,
169. Seale JV, Wood SA, Atkinson HC, Bate E, Lightman SL, Ingram CD, 2008.
Jessop DS, Harbuz MS. Gonadectomy reverses the sexually diergic 192. Tapp WN, Holaday JW, Natelson BH. Ultradian glucocorticoid rhythms
patterns of circadian and stress-induced hypothalamic-pituitary-adrenal in monkeys and rats continue during stress. Am J Physiol 247: R866-
axis activity in male and female rats. J Neuroendocrinol 16: 516-524, R871, 1984.
2004. 193. Tasker JG, Herman JP. Mechanisms of rapid glucocorticoid feedback
170. Seale JV, Wood SA, Atkinson HC, Harbuz MS, Lightman SL. Gonadal inhibition of the hypothalamic-pituitary-adrenal axis. Stress 14: 398-
steroid replacement reverses gonadectomy-induced changes in the cor- 406, 2011.
ticosterone pulse profile and stress-induced hypothalamic-pituitary- 194. Tousson E, Meissl H. Suprachiasmatic nuclei grafts restore the circadian
adrenal axis activity of male and female rats. J Neuroendocrinol 16: rhythm in the paraventricular nucleus of the hypothalamus. J Neurosci
989-998, 2004. 24: 2983-2988, 2004.
171. Seale JV, Wood SA, Atkinson HC, Harbuz MS, Lightman SL. Postnatal 195. Ulrich-Lai YM, Arnhold MM, Engeland WC. Adrenal splanchnic
masculinization alters the HPA axis phenotype in the adult female rat. innervation contributes to the diurnal rhythm of plasma corticosterone
J Physiol 563: 265-274, 2005. in rats by modulating adrenal sensitivity to ACTH. Am J Physiol Regul
172. Seale JV, Wood SA, Atkinson HC, Lightman SL, Harbuz MS. Orga- Integr Comp Physiol 290: R1128-R1135, 2006.
nizational role for testosterone and estrogen on adult hypothalamic- 196. Ulrich-Lai YM, Herman JP. Neural regulation of endocrine and auto-
pituitary-adrenal axis activity in the male rat. Endocrinology 146: 1973- nomic stress responses. Nat Rev Neurosci 10: 397-409, 2009.
1982, 2005. 197. Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue
173. Shanks N, Lightman SL. The maternal-neonatal neuro-immune inter- ovine hypothalamic peptide that stimulates secretion of corticotropin
face: Are there long-term implications for inflammatory or stress- and beta-endorphin. Science 213: 1394-1397, 1981.
related disease? J Clin Invest 108: 1567-1573, 2001. 198. Van Eekelen JA, De Kloet ER. Co-localization of brain corticosteroid
174. Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, receptors in the rat hippocampus. Prog Histochem Cytochem 26: 250-
Lightman SL. Early-life exposure to endotoxin alters hypothalamic- 258, 1992.
pituitary-adrenal function and predisposition to inflammation. Proc 199. Vrang N, Larsen PJ, Mikkelsen JD. Direct projection from the suprachi-
Natl Acad Sci U S A 97: 5645-5650, 2000. asmatic nucleus to hypophysiotrophic corticotropin-releasing factor
175. Son GH, Chung S, Choe HK, Kim HD, Baik SM, Lee H, Lee HW, immunoreactive cells in the paraventricular nucleus of the hypotha-
Choi S, Sun W, Kim H, Cho S, Lee KH, Kim K. Adrenal peripheral lamus demonstrated by means of Phaseolus vulgaris-leucoagglutinin
clock controls the autonomous circadian rhythm of glucocorticoid by tract tracing. Brain Res 684: 61-69, 1995.
causing rhythmic steroid production. Proc Natl Acad Sci U S A 105: 200. Waite EJ, McKenna M, Kershaw Y, Walker JJ, Cho K, Piggins HD,
20970-20975, 2008. Lightman SL. Ultradian corticosterone secretion is maintained in the
176. Song KH, Park JI, Lee MO, Soh J, Lee K, Choi HS. LH induces absence of circadian cues. Eur J Neurosci 36: 3142-3150, 2012.
orphan nuclear receptor Nur77 gene expression in testicular Leydig 201. Walker JJ, Spiga F, Waite E, Zhao Z, Kershaw Y, Terry JR, Lightman
cells. Endocrinology 142: 5116-5123, 2001. SL. The origin of glucocorticoid hormone oscillations. PLoS Biol 10:
177. Spencer RL, Miller AH, Moday H, Stein M, McEwen BS. Diurnal e1001341, 2012.
differences in basal and acute stress levels of type I and type II adrenal 202. Walker JJ, Terry JR, Lightman SL. Origin of ultradian pulsatility in the
steroid receptor activation in neural and immune tissues. Endocrinology hypothalamic-pituitary-adrenal axis. Proc Biol Sci R Soc 277: 1627-
133: 1941-1950, 1993. 1633, 2010.
178. Spiga F, Harrison LR, Wood SA, MacSweeney CP, Thomson FJ, Craig- 203. Wallberg AE, Flinn EM, Gustafsson JA, Wright AP. Recruitment of
head M, Grassie M, Lightman SL. Effect of the glucocorticoid receptor chromatin remodelling factors during gene activation via the glucocor-
antagonist Org 34850 on fast and delayed feedback of corticosterone ticoid receptor N-terminal domain. Biochem Soc Trans 28: 410-414,
release. J Endocrinol 196: 323-330, 2008. 2000.
179. Spiga F, Liu Y, Aguilera G, Lightman SL. Temporal effect of 204. Watts AG, Swanson LW. Efferent projections of the suprachiasmatic
adrenocorticotrophic hormone on adrenal glucocorticoid steroidoge- nucleus: II. Studies using retrograde transport of fluorescent dyes and
nesis: Involvement of the transducer of regulated cyclic AMP-response simultaneous peptide immunohistochemistry in the rat. J Comp Neurol
element-binding protein activity. J Neuroendocrinol 23: 136-142, 2011. 258: 230-252, 1987.
180. Spiga F, Waite EJ, Liu Y, Kershaw YM, Aguilera G, Lightman 205. Watts AG, Swanson LW, Sanchez-Watts G. Efferent projections of
SL. ACTH-Dependent Ultradian Rhythm of Corticosterone Secretion. the suprachiasmatic nucleus: I. Studies using anterograde transport of
Endocrinology 152: 1448-1457, 2011. Phaseolus vulgaris leucoagglutinin in the rat. J Comp Neurol 258: 204-
181. Stavreva DA, Wiench M, John S, Conway-Campbell BL, McKenna 229, 1987.
MA, Pooley JR, Johnson TA, Voss TC, Lightman SL, Hager GL. Ultra- 206. Watts AG, Tanimura S, Sanchez-Watts G. Corticotropin-releasing hor-
dian hormone stimulation induces glucocorticoid receptor-mediated mone and arginine vasopressin gene transcription in the hypothala-
pulses of gene transcription. Nat Cell Biol 11: 1093-1102, 2009. mic paraventricular nucleus of unstressed rats: Daily rhythms and
182. Stephan FK, Zucker I. Circadian rhythms in drinking behavior and their interactions with corticosterone. Endocrinology 145: 529-540,
locomotor activity of rats are eliminated by hypothalamic lesions. Proc 2004.
Natl Acad Sci U S A 69: 1583-1586, 1972. 207. Weitzman ED, Fukushima D, Nogeire C, Roffwarg H, Gallagher
183. Sternberg EM, Hill JM, Chrousos GP, Kamilaris T, Listwak SJ, TF, Hellman L. Twenty-four hour pattern of the episodic secretion
Gold PW, Wilder RL. Inflammatory mediator-induced hypothalamic- of cortisol in normal subjects. J Clin Endocrinol Metab 33: 14-22,
pituitary-adrenal axis activation is defective in streptococcal cell wall 1971.
arthritis-susceptible Lewis rats. Proc Natl Acad Sci U S A 86: 2374- 208. Welsh DK, Yoo SH, Liu AC, Takahashi JS, Kay SA. Biolumines-
2378, 1989. cence imaging of individual fibroblasts reveals persistent, indepen-
184. Sternberg EM, Young WS, 3rd, Bernardini R, Calogero AE, Chrousos dently phased circadian rhythms of clock gene expression. Curr Biol
GP, Gold PW, Wilder RL. A central nervous system defect in biosynthe- 14: 2289-2295, 2004.
sis of corticotropin-releasing hormone is associated with susceptibility 209. Widmaier EP, Dallman MF. The effects of corticotropin-releasing fac-
to streptococcal cell wall-induced arthritis in Lewis rats. Proc Natl Acad tor on adrenocorticotropin secretion from perifused pituitaries in vitro:
Sci U S A 86: 4771-4775, 1989. Rapid inhibition by glucocorticoids. Endocrinology 115: 2368-2374,
185. Stocco DM, Clark BJ. Regulation of the acute production of steroids in 1984.
steroidogenic cells. Endoc Rev 17: 221-244, 1996a. 210. Wilkinson CW, Shinsako J, Dallman MF. Return of pituitary-adrenal
186. Stocco DM, Clark BJ. Role of the steroidogenic acute regulatory protein function after adrenal enucleation or transplantation: Diurnal rhythms
(StAR) in steroidogenesis. Biochem Pharmacol 51: 197-205, 1996b. and responses to ether. Endocrinology 109: 162-169, 1981.
187. Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment 211. Windle RJ, Wood SA, Kershaw YM, Lightman SL, Ingram CD. Adap-
of the circadian clock in the liver by feeding. Science 291: 490-493, tive changes in basal and stress-induced HPA activity in lactating and
2001. post-lactating female rats. Endocrinology 154: 749-761, 2013.
188. Stratmann M, Schibler U. Properties, entrainment, and physiological 212. Windle RJ, Wood SA, Kershaw YM, Lightman SL, Ingram CD, Harbuz
functions of mammalian peripheral oscillators. J Biol Rhythms 21: 494- MS. Increased corticosterone pulse frequency during adjuvant-induced
506, 2006. arthritis and its relationship to alterations in stress responsiveness.
189. Sugawara T, Holt JA, Kiriakidou M, Strauss JF, 3rd. Steroidogenic J Neuroendocrinol 13: 905-911, 2001.
factor 1-dependent promoter activity of the human steroidogenic acute 213. Windle RJ, Wood SA, Lightman SL, Ingram CD. The pulsatile char-
regulatory protein (StAR) gene. Biochemistry 35: 9052-9059, 1996. acteristics of hypothalamo-pituitary-adrenal activity in female Lewis

Volume 4, July 2014 1297


HPA Axis-Rhythms Comprehensive Physiology

and Fischer 344 rats and its relationship to differential stress responses. interaction and nuclear translocation of the glucocorticoid receptor in
Endocrinology 139: 4044-4052, 1998. mammalian cells. J Biol Chem 280: 4609-4616, 2005.
214. Windle RJ, Wood SA, Shanks N, Lightman SL, Ingram CD. Ultradian 216. Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block
rhythm of basal corticosterone release in the female rat: Dynamic inter- GD, Sakaki Y, Menaker M, Tei H. Resetting central and peripheral
action with the response to acute stress. Endocrinology 139: 443-450, circadian oscillators in transgenic rats. Science 288: 682-685, 2000.
1998. 217. Zazopoulos E, Lalli E, Stocco DM, Sassone-Corsi P. DNA binding and
215. Wochnik GM, Ruegg J, Abel GA, Schmidt U, Holsboer F, Rein transcriptional repression by DAX-1 blocks steroidogenesis. Nature
T. FK506-binding proteins 51 and 52 differentially regulate dynein 390: 311-315, 1997.

1298 Volume 4, July 2014

You might also like