Comprehensive Physiology Wiley Online Library

HPA Axis‐Rhythms

Full Article on Wiley Online Library



Abstract

The hypothalamic‐pituitary‐adrenal (HPA) axis regulates circulating levels of glucocorticoid hormones, 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. © 2014 American Physiological Society. Compr Physiol 4:1273‐1298, 2014.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. 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 (201).
Figure 2. 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).
Figure 3. 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 (201).
Figure 4. Figure 4. An ultradian pattern underlies all the components of the HPA axis. (A) Pulsatile release of CRH in the rat median eminence. CRH was measured in perfusate from push‐pull cannulas implanted in the rat median eminence; samples were collected at 5 min intervals from an unanesthetized male rat under basal conditions. Reproduced with permission from (84). (B) ACTH levels in rat plasma samples collected every 2 min from the jugular vein via an indwelling cannula. Reproduced with permission from (27). (C) Corticosterone levels in rat blood samples collected every 5 min using an automated blood sampling system. Reproduced with permission from (202). Note that in the rat, CRH pulse frequency is higher (∼3 pulses/h) than the near‐hourly oscillation in ACTH and corticosterone.
Figure 5. Figure 5. Human cortisol and ACTH dynamics. Ultradian ACTH and cortisol oscillations in a healthy man are tightly correlated with ACTH leading cortisol. ACTH and cortisol were measured in blood samples collected at 10 min intervals using an automated blood sampling system. Light was off between 2300 and 0700. Adapted and reproduced with permission from (73).
Figure 6. 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 (203).
Figure 7. 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 (202).
Figure 8. 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).
Figure 9. 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 corticosterone 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 (213). In all experiments, corticosterone was measured in blood samples collected at 10 min intervals from freely behaving rats. Shaded regions indicate the dark phase.
Figure 10. Figure 10. Stress responsiveness is dependent on the phase of the ultradian corticosterone rhythm. Corticosterone response in rats exposed to a noise stress (10 min, 114 dB); blood samples were collected every 10 min using an automated blood sampling system. Rats stressed during the rising phase of an endogenous corticosterone pulse (A) show much greater corticosterone responses than animals stressed during the falling phase (B). Reproduced with permission from (215).
Figure 11. Figure 11. Ultradian rhythm of hippocampal free corticosterone measured by in vivo microdialysis 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).
Figure 12. Figure 12. Ultradian corticosterone pulses induce pulsatile GR‐GRE interactions and pulsatile gene transcription. (A) Real time single cell imaging of GFP‐labeled GR loading at an engineered array of GREs in the MCF‐7 (3617) mouse cell line. Each pulse of corticosterone causes a “wave” of GR‐GRE binding (fluorescent green points) that is rapidly reversed (GR‐GRE dissociation) following hormone withdrawal (washout). (B) Dependence of transcriptional dynamics on the pattern of hormone stimulation. Ultradian corticosterone treatment results in pulsatile transcription of the Period 1 gene (Per1) (black), whereas constant corticosterone induces sustained transcription (grey). Levels of Per1 primary transcript were measured in the 3134 cell line by real‐time quantitative PCR. Data reproduced with permission from (182).
Figure 13. Figure 13. Corticosterone ultradian rhythm induces pulsatile GR activation and GR‐mediated gene transcription 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).


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 (201).


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).


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 (201).


Figure 4. An ultradian pattern underlies all the components of the HPA axis. (A) Pulsatile release of CRH in the rat median eminence. CRH was measured in perfusate from push‐pull cannulas implanted in the rat median eminence; samples were collected at 5 min intervals from an unanesthetized male rat under basal conditions. Reproduced with permission from (84). (B) ACTH levels in rat plasma samples collected every 2 min from the jugular vein via an indwelling cannula. Reproduced with permission from (27). (C) Corticosterone levels in rat blood samples collected every 5 min using an automated blood sampling system. Reproduced with permission from (202). Note that in the rat, CRH pulse frequency is higher (∼3 pulses/h) than the near‐hourly oscillation in ACTH and corticosterone.


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


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 (203).


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 (202).


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).


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 corticosterone 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 (213). In all experiments, corticosterone was measured in blood samples collected at 10 min intervals from freely behaving rats. Shaded regions indicate the dark phase.


Figure 10. Stress responsiveness is dependent on the phase of the ultradian corticosterone rhythm. Corticosterone response in rats exposed to a noise stress (10 min, 114 dB); blood samples were collected every 10 min using an automated blood sampling system. Rats stressed during the rising phase of an endogenous corticosterone pulse (A) show much greater corticosterone responses than animals stressed during the falling phase (B). Reproduced with permission from (215).


Figure 11. Ultradian rhythm of hippocampal free corticosterone measured by in vivo microdialysis 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).


Figure 12. Ultradian corticosterone pulses induce pulsatile GR‐GRE interactions and pulsatile gene transcription. (A) Real time single cell imaging of GFP‐labeled GR loading at an engineered array of GREs in the MCF‐7 (3617) mouse cell line. Each pulse of corticosterone causes a “wave” of GR‐GRE binding (fluorescent green points) that is rapidly reversed (GR‐GRE dissociation) following hormone withdrawal (washout). (B) Dependence of transcriptional dynamics on the pattern of hormone stimulation. Ultradian corticosterone treatment results in pulsatile transcription of the Period 1 gene (Per1) (black), whereas constant corticosterone induces sustained transcription (grey). Levels of Per1 primary transcript were measured in the 3134 cell line by real‐time quantitative PCR. Data reproduced with permission from (182).


Figure 13. Corticosterone ultradian rhythm induces pulsatile GR activation and GR‐mediated gene transcription 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).
References
 1. Abe K , Kroning J , Greer MA , Critchlow V . Effects of destruction of the suprachiasmatic nuclei on the circadian rhythms in plasma corticosterone, body temperature, feeding and plasma thyrotropin. Neuroendocrinology 29: 119‐131, 1979.
 2. Aguilera G , Jessop DS , Harbuz MS , Kiss A , Lightman SL . Differential regulation of hypothalamic pituitary corticotropin releasing hormone receptors during development of adjuvant‐induced arthritis in the rat. J Endocrinol 153: 185‐191, 1997.
 3. Ahima RS , Harlan RE . Charting of type II glucocorticoid receptor‐like immunoreactivity in the rat central nervous system. Neuroscience 39: 579‐604, 1990.
 4. Akana SF , Cascio CS , Du JZ , Levin N , Dallman MF . Reset of feedback in the adrenocortical system: An apparent shift in sensitivity of adrenocorticotropin to inhibition by corticosterone between morning and evening. Endocrinology 119: 2325‐2332, 1986.
 5. Alexander LD , Sander LD . Vasoactive intestinal peptide stimulates ACTH and corticosterone release after injection into the PVN. Regul Pept 51: 221‐227, 1994.
 6. Antoni FA . Hypothalamic control of adrenocorticotropin secretion: Advances since the discovery of 41‐residue corticotropin‐releasing factor. Endoc Rev 7: 351‐378, 1986.
 7. Antoni FA , Holmes MC , Makara GB , Karteszi M , Laszlo FA . Evidence that the effects of arginine‐8‐vasopressin (AVP) on pituitary corticotropin (ACTH) release are mediated by a novel type of receptor. Peptides 5: 519‐522, 1984.
 8. Apostolakis EM , Longo LD , Veldhuis JD , Yellon SM . Dissociation of pulsatile cortisol and adrenocorticotropin secretion in fetal sheep during late gestation. Endocrinology 130: 2571‐2578, 1992.
 9. Arakane F , King SR , Du Y , Kallen CB , Walsh LP , Watari H , Stocco DM , Strauss JF, II . Phosphorylation of steroidogenic acute regulatory protein (StAR) modulates its steroidogenic activity. J Biol Chem 272: 32656‐32662, 1997.
 10. Arriza JL , Simerly RB , Swanson LW , Evans RM . The neuronal mineralocorticoid receptor as a mediator of glucocorticoid response. Neuron 1: 887‐900, 1988.
 11. Artemenko IP , Zhao D , Hales DB , Hales KH , Jefcoate CR . Mitochondrial processing of newly synthesized steroidogenic acute regulatory protein (StAR), but not total StAR, mediates cholesterol transfer to cytochrome P450 side chain cleavage enzyme in adrenal cells. J Biol 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 International Society of Psychoneuroendocrinology (ISPNE). Leiden, The Netherlands.: 2013.
 14. Bhake RC , Leendertz JA , Linthorst AC , Lightman SL . Automated 24‐hours sampling of subcutaneous tissue free cortisol in humans. J Med Eng Technol 37: 180‐184, 2013.
 15. Bingaman EW , Magnuson DJ , Gray TS , Handa RJ . Androgen inhibits the increases in hypothalamic corticotropin‐releasing hormone (CRH) and CRH‐immunoreactivity following gonadectomy. Neuroendocrinology 59: 228‐234, 1994.
 16. Bittman EL , Doherty L , Huang L , Paroskie A . Period gene expression in mouse endocrine tissues. Am J Physiol Regul Integr Comp Physiol 285: R561‐R569, 2003.
 17. Buijs RM , Kalsbeek A , van der Woude TP , van Heerikhuize JJ , Shinn S . Suprachiasmatic nucleus lesion increases corticosterone secretion. Am J Physiol 264: R1186‐R1192, 1993.
 18. Buijs RM , Van Eden CG . The integration of stress by the hypothalamus, amygdala and prefrontal cortex: Balance between the autonomic nervous system and the neuroendocrine system. Prog Brain Res 126: 117‐132, 2000.
 19. Buijs RM , Wortel J , Van Heerikhuize JJ , Feenstra MG , Ter Horst GJ , Romijn HJ , Kalsbeek A . Anatomical and functional demonstration of a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway. Eur J Neurosci 11: 1535‐1544, 1999.
 20. Bunger MK , Wilsbacher LD , Moran SM , Clendenin C , Radcliffe LA , Hogenesch JB , Simon MC , Takahashi JS , Bradfield CA . Mop3 is an essential component of the master circadian pacemaker in mammals. Cell 103: 1009‐1017, 2000.
 21. Bush IE . Species differences in adrenocortical secretion. J Endocrinol 9: 95‐100, 1953.
 22. Camacho‐Arroyo I , Neri‐Gomez T , Gonzalez‐Arenas A , Guerra‐Araiza C . Changes in the content of steroid receptor coactivator‐1 and silencing mediator for retinoid and thyroid hormone receptors in the rat brain during the estrous cycle. J Steroid Biochem Mol Biol 94: 267‐272, 2005.
 23. Cameron A , Henley D , Carrell R , Zhou A , Clarke A , Lightman S . Temperature‐responsive release of cortisol from its binding globulin: A protein thermocouple. J Clin Endocrinol Metab 95: 4689‐4695, 2010.
 24. Caraty A , Grino M , Locatelli A , Oliver C . Secretion of corticotropin releasing factor (CRF) and vasopressin (AVP) into the hypophysial portal blood of conscious, unrestrained rams. Biochem Biophys Res Commun 155: 841‐849, 1988.
 25. Carnes M , Brownfield MS , Kalin NH , Lent S , Barksdale CM . Episodic secretion of ACTH in rats. Peptides 7: 219‐223, 1986.
 26. Carnes M , Kalin NH , Lent SJ , Barksdale CM , Brownfield MS . Pulsatile ACTH secretion: Variation with time of day and relationship to cortisol. Peptides 9: 325‐331, 1988.
 27. Carnes M , Lent S , Feyzi J , Hazel D . Plasma adrenocorticotropic hormone in the rat demonstrates three different rhythms within 24 h. Neuroendocrinology 50: 17‐25, 1989.
 28. Carnes M , Lent SJ , Erisman S , Barksdale C , Feyzi J . Immunoneutralization of corticotropin‐releasing hormone prevents the diurnal surge of ACTH. Life Sci 45: 1049‐1056, 1989.
 29. Carnes M , Lent SJ , Erisman S , Feyzi J . Changes in mean plasma ACTH reflect changes in amplitude and frequency of secretory pulses. Life Sci 43: 1785‐1790, 1988.
 30. Chalmers DT , Lovenberg TW , De Souza EB . Localization of novel corticotropin‐releasing factor receptor (CRF2) mRNA expression to specific subcortical nuclei in rat brain: Comparison with CRF1 receptor mRNA expression. J Neurosci 15: 6340‐6350, 1995.
 31. Chaudhary LR , Stocco DM . Effect of different steroidogenic stimuli on protein phosphorylation and steroidogenesis in MA‐10 mouse Leydig tumor cells. Biochim Biophys Acta 1094: 175‐184, 1991.
 32. Cherrington AD . Banting Lecture 1997. Control of glucose uptake and release by the liver in vivo. Diabetes 48: 1198‐1214, 1999.
 33. Chikanza IC , Chrousos G , Panayi GS . Abnormal neuroendocrine immune communications in patients with rheumatoid arthritis. Eur J Clin Invest 22: 635‐637, 1992.
 34. Chowdrey HS , Larsen PJ , Harbuz MS , Jessop DS , Aguilera G , Eckland DJ , Lightman SL . Evidence for arginine vasopressin as the primary activator of the HPA axis during adjuvant‐induced arthritis. Br J Pharmacol 116: 2417‐2424, 1995.
 35. Chrousos GP . The hypothalamic‐pituitary‐adrenal axis and immune‐mediated inflammation. N Engl J Med 332: 1351‐1362, 1995.
 36. Churchill PF , Kimura T . Topological studies of cytochromes P‐450scc and P‐45011 beta in bovine adrenocortical inner mitochondrial membranes. Effects of controlled tryptic digestion. J Biol Chem 254: 10443‐10448, 1979.
 37. Clark RG , Chambers G , Lewin J , Robinson IC . Automated repetitive microsampling of blood: Growth hormone profiles in conscious male rats. J Endocrinol 111: 27‐35, 1986.
 38. Conway‐Campbell BL , George CL , Pooley JR , Knight DM , Norman MR , Hager GL , Lightman SL . The HSP90 molecular chaperone cycle regulates cyclical transcriptional dynamics of the glucocorticoid receptor and its coregulatory molecules CBP/p300 during ultradian ligand treatment. Mol Endocrinol 25: 944‐954, 2011.
 39. Conway‐Campbell BL , McKenna MA , Wiles CC , Atkinson HC , de Kloet ER , Lightman SL . Proteasome‐dependent down‐regulation of activated nuclear hippocampal glucocorticoid receptors determines dynamic responses to corticosterone. Endocrinology 148: 5470‐5477, 2007.
 40. Conway‐Campbell BL , Pooley JR , Hager GL , Lightman SL . Molecular dynamics of ultradian glucocorticoid receptor action. Mol Cell Endocrinol 348: 383‐393, 2012.
 41. Conway‐Campbell BL , Sarabdjitsingh RA , McKenna MA , Pooley JR , Kershaw YM , Meijer OC , De Kloet ER , Lightman SL . Glucocorticoid ultradian rhythmicity directs cyclical gene pulsing of the clock gene period 1 in rat hippocampus. J Neuroendocrinol 22: 1093‐1100, 2010.
 42. Dallman MF , Akana SF , Cascio CS , Darlington DN , Jacobson L , Levin N . Regulation of ACTH secretion: Variations on a theme of B. Recent Prog Horm Res 43: 113‐173, 1987.
 43. Dallman MF , Akana SF , Jacobson L , Levin N , Cascio CS , Shinsako J . Characterization of corticosterone feedback regulation of ACTH secretion. Ann N Y Acad Sci 512: 402‐414, 1987.
 44. Dallman MF , Engeland WC , Rose JC , Wilkinson CW , Shinsako J , Siedenburg F . Nycthemeral rhythm in adrenal responsiveness to ACTH. Am J Physiol 235: R210‐R218, 1978.
 45. Dallman MF , Jones MT . Corticosteroid feedback control of ACTH secretion: Effect of stress‐induced corticosterone ssecretion on subsequent stress responses in the rat. Endocrinology 92: 1367‐1375, 1973.
 46. Dallmann R , Touma C , Palme R , Albrecht U , Steinlechner S . Impaired daily glucocorticoid rhythm in Per1 (Brd) mice. J Comp Physiol [A] 192: 769‐775, 2006.
 47. De Kloet ER . Hormones and the stressed brain. Ann N Y Acad Sci 1018: 1‐15, 2004.
 48. de Kloet ER . Steroids, stability and stress. Front Neuroendocrinol 16: 416‐425, 1995.
 49. de Kloet ER . Stress in the brain. Eur J Pharmacol 405: 187‐198, 2000.
 50. De Kloet ER , Vreugdenhil E , Oitzl MS , Joels M . Brain corticosteroid receptor balance in health and disease. Endoc Rev 19: 269‐301, 1998.
 51. Di S , Malcher‐Lopes R , Halmos KC , Tasker JG . Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: A fast feedback mechanism. J Neurosci 23: 4850‐4857, 2003.
 52. Droste SK , Collins A , Lightman SL , Linthorst AC , Reul JM . Distinct, time‐dependent effects of voluntary exercise on circadian and ultradian rhythms and stress responses of free corticosterone in the rat hippocampus. Endocrinology 150: 4170‐4179, 2009.
 53. Droste SK , de Groote L , Atkinson HC , Lightman SL , Reul JM , Linthorst AC . Corticosterone levels in the brain show a distinct ultradian rhythm but a delayed response to forced swim stress. Endocrinology 149: 3244‐3253, 2008.
 54. Droste SK , de Groote L , Lightman SL , Reul JM , Linthorst AC . The ultradian and circadian rhythms of free corticosterone in the brain are not affected by gender: An in vivo microdialysis study in Wistar rats. J Neuroendocrinol 21: 132‐140, 2009.
 55. Engler D , Pham T , Fullerton MJ , Clarke IJ , Funder JW . Evidence for an ultradian secretion of adrenocorticotropin, beta‐endorphin and alpha‐melanocyte‐stimulating hormone by the ovine anterior and intermediate pituitary. Neuroendocrinology 49: 349‐360, 1989.
 56. Engler D , Pham T , Liu JP , Fullerton MJ , Clarke IJ , Funder JW . Studies of the regulation of the hypothalamic‐pituitary‐adrenal axis in sheep with hypothalamic‐pituitary disconnection. II. Evidence for in vivo ultradian hypersecretion of proopiomelanocortin peptides by the isolated anterior and intermediate pituitary. Endocrinology 127: 1956‐1966, 1990.
 57. Evanson NK , Tasker JG , Hill MN , Hillard CJ , Herman JP . Fast feedback inhibition of the HPA axis by glucocorticoids is mediated by endocannabinoid signaling. Endocrinology 151: 4811‐4819, 2010.
 58. Fahrenkrug J , Hannibal J , Georg B . Diurnal rhythmicity of the canonical clock genes Per1, Per2 and Bmal1 in the rat adrenal gland is unaltered after hypophysectomy. J Neuroendocrinol 20: 323‐329, 2008.
 59. Fulkerson WJ . Synchronous episodic release of cortisol in the sheep. J Endocrinol 79: 131‐132, 1978.
 60. Gala RR , Westphal U . Corticosteroid‐binding globulin in the rat: Studies on the sex difference. Endocrinology 77: 841‐851, 1965.
 61. Gekakis N , Staknis D , Nguyen HB , Davis FC , Wilsbacher LD , King DP , Takahashi JS , Weitz CJ . Role of the CLOCK protein in the mammalian circadian mechanism. Science 280: 1564‐1569, 1998.
 62. Gillies GE , Linton EA , Lowry PJ . Corticotropin releasing activity of the new CRF is potentiated several times by vasopressin. Nature 299: 355‐357, 1982.
 63. Girotti M , Pace TW , Gaylord RI , Rubin BA , Herman JP , Spencer RL . Habituation to repeated restraint stress is associated with lack of stress‐induced c‐fos expression in primary sensory processing areas of the rat brain. Neuroscience 138: 1067‐1081, 2006.
 64. Graham ES , Littlewood P , Turnbull Y , Mercer JG , Morgan PJ , Barrett P . Neuromedin‐U is regulated by the circadian clock in the SCN of the mouse. Eur J Neurosci 21: 814‐819, 2005.
 65. Haller J , Halasz J , Mikics E , Kruk MR , Makara GB . Ultradian corticosterone rhythm and the propensity to behave aggressively in male rats. J Neuroendocrinol 12: 937‐940, 2000.
 66. Haller J , Millar S , van de Schraaf J , de Kloet RE , Kruk MR . The active phase‐related increase in corticosterone and aggression are linked. J Neuroendocrinol 12: 431‐436, 2000.
 67. Handa RJ , Weiser MJ . Gonadal steroid hormones and the hypothalamo‐pituitary‐adrenal axis. Front Neuroendocrinol 35: 197‐220, 2014.
 68. Harbuz MS , Rees RG , Eckland D , Jessop DS , Brewerton D , Lightman SL . Paradoxical responses of hypothalamic corticotropin‐releasing factor (CRF) messenger ribonucleic acid (mRNA) and CRF‐41 peptide and adenohypophysial proopiomelanocortin mRNA during chronic inflammatory stress. Endocrinology 130: 1394‐1400, 1992.
 69. Harbuz MS , Rees RG , Lightman SL . HPA axis responses to acute stress and adrenalectomy during adjuvant‐induced arthritis in the rat. Am J Physiol 264: R179‐R185, 1993.
 70. Hastings M , Maywood ES . Circadian clocks in the mammalian brain. BioEssays 22: 23‐31, 2000.
 71. Hastings M , O'Neill JS , Maywood ES . Circadian clocks: Regulators of endocrine and metabolic rhythms. J Endocrinol 195: 187‐198, 2007.
 72. Hendrickson AE , Wagoner N , Cowan WM . An autoradiographic and electron microscopic study of retino‐hypothalamic connections. Z Zellforsch Mikrosk Anat 135: 1‐26, 1972.
 73. Henley DE , Leendertz JA , Russell GM , Wood SA , Taheri S , Woltersdorf WW , Lightman SL . Development of an automated blood sampling system for use in humans. J Med Eng Technol 33: 199‐208, 2009.
 74. Hill MN , Tasker JG . Endocannabinoid signaling, glucocorticoid‐mediated negative feedback, and regulation of the hypothalamic‐pituitary‐adrenal axis. Neuroscience 204: 5‐16, 2012.
 75. Hinz B , Hirschelmann R . Rapid non‐genomic feedback effects of glucocorticoids on CRF‐induced ACTH secretion in rats. Pharm Res 17: 1273‐1277, 2000.
 76. Holaday JW , Martinez HM , Natelson BH . Synchronized ultradian cortisol rhythms in monkeys: Persistence during corticotropin infusion. Science 198: 56‐58, 1977.
 77. Hsu BR , Kuhn RW . The role of the adrenal in generating the diurnal variation in circulating levels of corticosteroid‐binding globulin in the rat. Endocrinology 122: 421‐426, 1988.
 78. Hubler TR , Scammell JG . Intronic hormone response elements mediate regulation of FKBP5 by progestins and glucocorticoids. Cell Stress Chaperones 9: 243‐252, 2004.
 79. Hugin‐Flores ME , Steimer T , Aubert ML , Schulz P . Mineralo‐ and glucocorticoid receptor mrnas are differently regulated by corticosterone in the rat hippocampus and anterior pituitary. Neuroendocrinology 79: 174‐184, 2004.
 80. Hum DW , Miller WL . Transcriptional regulation of human genes for steroidogenic enzymes. Clin Chem 39: 333‐340, 1993.
 81. Ishida A , Mutoh T , Ueyama T , Bando H , Masubuchi S , Nakahara D , Tsujimoto G , Okamura H . Light activates the adrenal gland: Timing of gene expression and glucocorticoid release. Cell Metab 2: 297‐307, 2005.
 82. Iwasaki‐Sekino A , Mano‐Otagiri A , Ohata H , Yamauchi N , Shibasaki T . Gender differences in corticotropin and corticosterone secretion and corticotropin‐releasing factor mRNA expression in the paraventricular nucleus of the hypothalamus and the central nucleus of the amygdala in response to footshock stress or psychological stress in rats. Psychoneuroendocrinology 34: 226‐237, 2009.
 83. Ixart G , Barbanel G , Nouguier‐Soule J , Assenmacher I . A quantitative study of the pulsatile parameters of CRH‐41 secretion in unanesthetized free‐moving rats. Exp Brain Res 87: 153‐158, 1991.
 84. Ixart G , Siaud P , Barbanel G , Mekaouche M , Givalois L , Assenmacher I . Circadian variations in the amplitude of corticotropin‐releasing hormone 41 (CRH41) episodic release measured in vivo in male rats: Correlations with diurnal fluctuations in hypothalamic and median eminence CRH41 contents. J Biol Rhythms 8: 297‐309, 1993.
 85. Ixart G , Siaud P , Mekaouche M , Barbanel G , Givalois L , Assenmacher I . Short‐term but not long‐term adrenalectomy modulates amplitude and frequency of the CRH41 episodic release in push‐pull cannulated median eminence of free‐moving rats. Brain Res 658: 185‐191, 1994.
 86. Jasper MS , Engeland WC . Synchronous ultradian rhythms in adrenocortical secretion detected by microdialysis in awake rats. Am J Physiol 261: R1257‐R1268, 1991.
 87. Jasper MS , Engeland WC . Splanchnic neural activity modulates ultradian and circadian rhythms in adrenocortical secretion in awake rats. Neuroendocrinology 59: 97‐109, 1994.
 88. Jasper MS , Engeland WC . Splanchnicotomy increases adrenal sensitivity to ACTH in nonstressed rats. Am J Physiol 273: E363‐E368, 1997.
 89. Joels M , Urban IJ . The effect of microiontophoretically applied vasopressin and oxytocin on single neurones in the septum and dorsal hippocampus of the rat. Neurosci Lett 33: 79‐84, 1982.
 90. Jones MT , Brush FR , Neame RL . Characteristics of fast feedback control of corticotrophin release by corticosteroids. J Endocrinol 55: 489‐497, 1972.
 91. Jones MT , Hillhouse EW , Burden JL . Dynamics and mechanics of corticosteroid feedback at the hypothalamus and anterior pituitary gland. J Endocrinol 73: 405‐417, 1977.
 92. Jones MT , Tiptaft EM , Brush FR , Fergusson DA , Neame RL . Evidence for dual corticosteroid‐receptor mechanisms in the feedback control of adrenocorticotrophin secretion. J Endocrinol 60: 223‐233, 1974.
 93. Kalsbeek A , Buijs RM , van Heerikhuize JJ , Arts M , van der Woude TP . Vasopressin‐containing neurons of the suprachiasmatic nuclei inhibit corticosterone release. Brain Res 580: 62‐67, 1992.
 94. Kalsbeek A , Fliers E , Hofman MA , Swaab DF , Buijs RM . Vasopressin and the output of the hypothalamic biological clock. J Neuroendocrinol 22: 362‐372, 2010.
 95. Kalsbeek A , van der Spek R , Lei J , Endert E , Buijs RM , Fliers E . Circadian rhythms in the hypothalamo‐pituitary‐adrenal (HPA) axis. Mol Cell Endocrinol 349: 20‐29, 2012.
 96. Kalsbeek A , van der Vliet J , Buijs RM . Decrease of endogenous vasopressin release necessary for expression of the circadian rise in plasma corticosterone: A reverse microdialysis study. J Neuroendocrinol 8: 299‐307, 1996.
 97. Kalsbeek A , van Heerikhuize JJ , Wortel J , Buijs RM . A diurnal rhythm of stimulatory input to the hypothalamo‐pituitary‐adrenal system as revealed by timed intrahypothalamic administration of the vasopressin V1 antagonist. J Neurosci 16: 5555‐5565, 1996.
 98. Kaneko M , Hiroshige T , Shinsako J , Dallman MF . Diurnal changes in amplification of hormone rhythms in the adrenocortical system. Am J Physiol 239: R309‐R316, 1980.
 99. Karssen AM , Meijer OC , van der Sandt IC , Lucassen PJ , de Lange EC , de Boer AG , de Kloet ER . Multidrug resistance P‐glycoprotein hampers the access of cortisol but not of corticosterone to mouse and human brain. Endocrinology 142: 2686‐2694, 2001.
 100. Karst H , Berger S , Erdmann G , Schutz G , Joels M . Metaplasticity of amygdalar responses to the stress hormone corticosterone. Proc Natl Acad Sci U S A 107: 14449‐14454, 2010.
 101. Karst H , Berger S , Turiault M , Tronche F , Schutz G , Joels M . Mineralocorticoid receptors are indispensable for nongenomic modulation of hippocampal glutamate transmission by corticosterone. Proc Natl Acad Sci U S A 102: 19204‐19207, 2005.
 102. Kasai M , Kannan H , Ueta Y , Osaka T , Inenaga K , Yamashita H . Effects of iontophoretically applied cortisol on tuberoinfundibular neurons in hypothalamic paraventricular nucleus of anesthetized rats. Neurosci Lett 87: 35‐40, 1988.
 103. Khan MS , Aden D , Rosner W . Human corticosteroid binding globulin is secreted by a hepatoma‐derived cell line. J Steroid Biochem 20: 677‐678, 1984.
 104. King DP , Zhao Y , Sangoram AM , Wilsbacher LD , Tanaka M , Antoch MP , Steeves TD , Vitaterna MH , Kornhauser JM , Lowrey PL , Turek FW , Takahashi JS . Positional cloning of the mouse circadian clock gene. Cell 89: 641‐653, 1997.
 105. Kino T , Nordeen SK , Chrousos GP . Conditional modulation of glucocorticoid receptor activities by CREB‐binding protein (CBP) and p300. J Steroid Biochem Mol Biol 70: 15‐25, 1999.
 106. Kitchener P , Di Blasi F , Borrelli E , Piazza PV . Differences between brain structures in nuclear translocation and DNA binding of the glucocorticoid receptor during stress and the circadian cycle. Eur J Neurosci 19: 1837‐1846, 2004.
 107. Kow LM , Pfaff DW . Vasopressin excites ventromedial hypothalamic glucose‐responsive neurons in vitro. Physiol Behav 37: 153‐158, 1986.
 108. Kruk MR , Halasz J , Meelis W , Haller J . Fast positive feedback between the adrenocortical stress response and a brain mechanism involved in aggressive behavior. Behav Neurosci 118: 1062‐1070, 2004.
 109. Lamia KA , Storch KF , Weitz CJ . Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci U S A 105: 15172‐15177, 2008.
 110.Le Martelot G , Claudel T , Gatfield D , Schaad O , Kornmann B , Lo Sasso G , Moschetta A , Schibler U . REV‐ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol 7: e1000181, 2009.
 111. Leliavski A , Shostak A , Husse J , Oster H . Impaired glucocorticoid production and response to stress in arntl‐deficient male mice. Endocrinology 155: 133‐142, 2014.
 112. Levi F , Schibler U . Circadian rhythms: Mechanisms and therapeutic implications. Annu Rev Pharmacol Toxicol 47: 593‐628, 2007.
 113. Lewis JG , Bagley CJ , Elder PA , Bachmann AW , Torpy DJ . Plasma free cortisol fraction reflects levels of functioning corticosteroid‐binding globulin. Clin Chim Acta 359: 189‐194, 2005.
 114. Lewis JG , Mopert B , Shand BI , Doogue MP , Soule SG , Frampton CM , Elder PA . Plasma variation of corticosteroid‐binding globulin and sex hormone‐binding globulin. Horm Metab Res 38: 241‐245, 2006.
 115. Li TZ , Wang CA , Chen YZ . [The rapid effect of the iontophoretically applied cortisol on unit activity of neurons in three brain areas in rats]. Sheng li xue bao : [Acta physiologica Sinica] 43: 280‐285, 1991.
 116. Lightman SL , Conway‐Campbell BL . The crucial role of pulsatile activity of the HPA axis for continuous dynamic equilibration. Nat Rev Neurosci 11: 710‐718, 2010.
 117. Lightman SL , Windle RJ , Julian MD , Harbuz MS , Shanks N , Wood SA , Kershaw YM , Ingram CD . Significance of pulsatility in the HPA axis. Novartis Found Symp 227: 244‐257; discussion 257‐260, 2000.
 118. Lin D , Sugawara T , Strauss JF, III , Clark BJ , Stocco DM , Saenger P , Rogol A , Miller WL . Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science 267: 1828‐1831, 1995.
 119. Liu Y , Smith LI , Huang V , Poon V , Coello A , Olah M , Spiga F , Lightman SL , Aguilera G . Transcriptional regulation of episodic glucocorticoid secretion. Mol Cell Endocrinol 371: 62‐70, 2013.
 120. Loh DH , Abad C , Colwell CS , Waschek JA . Vasoactive intestinal peptide is critical for circadian regulation of glucocorticoids. Neuroendocrinology 88: 246‐255, 2008.
 121. Macfarlane DP , Forbes S , Walker BR . Glucocorticoids and fatty acid metabolism in humans: Fuelling fat redistribution in the metabolic syndrome. J Endocrinol 197: 189‐204, 2008.
 122. Mahmoud SN , Scaccianoce S , Scraggs PR , Nicholson SA , Gillham B , Jones MT . Characteristics of corticosteroid inhibition of adrenocorticotrophin release from the anterior pituitary gland of the rat. J Endocrinol 102: 33‐42, 1984.
 123. Mangelsdorf DJ , Thummel C , Beato M , Herrlich P , Schutz G , Umesono K , Blumberg B , Kastner P , Mark M , Chambon P , Evans RM . The nuclear receptor superfamily: The second decade. Cell 83: 835‐839, 1995.
 124. Marcheva B , Ramsey KM , Buhr ED , Kobayashi Y , Su H , Ko CH , Ivanova G , Omura C , Mo S , Vitaterna MH , Lopez JP , Philipson LH , Bradfield CA , Crosby SD , JeBailey L , Wang X , Takahashi JS , Bass J . Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466: 627‐631, 2010.
 125. Mataradze GD , Kurabekova RM , Rozen VB . The role of sex steroids in the formation of sex‐differentiated concentrations of corticosteroid‐binding globulin in rats. J Endocrinol 132: 235‐240, 1992.
 126. Maywood ES , O'Neill JS , Chesham JE , Hastings MH . Minireview: The circadian clockwork of the suprachiasmatic nuclei–analysis of a cellular oscillator that drives endocrine rhythms. Endocrinology 148: 5624‐5634, 2007.
 127. McDearmon EL , Patel KN , Ko CH , Walisser JA , Schook AC , Chong JL , Wilsbacher LD , Song EJ , Hong HK , Bradfield CA , Takahashi JS . Dissecting the functions of the mammalian clock protein BMAL1 by tissue‐specific rescue in mice. Science 314: 1304‐1308, 2006.
 128. McEwen BS . Physiology and neurobiology of stress and adaptation: Central role of the brain. Physiol Rev 87: 873‐904, 2007.
 129. Meijer OC , Steenbergen PJ , De Kloet ER . Differential expression and regional distribution of steroid receptor coactivators SRC‐1 and SRC‐2 in brain and pituitary. Endocrinology 141: 2192‐2199, 2000.
 130. Merriam GR , Wachter KW . Algorithms for the study of episodic hormone secretion. Am J Physiol 243: E310‐E318, 1982.
 131. Mershon JL , Sehlhorst CS , Rebar RW , Liu JH . Evidence of a corticotropin‐releasing hormone pulse generator in the macaque hypothalamus. Endocrinology 130: 2991‐2996, 1992.
 132. Metherell LA , Chapple JP , Cooray S , David A , Becker C , Ruschendorf F , Naville D , Begeot M , Khoo B , Nurnberg P , Huebner A , Cheetham ME , Clark AJ . Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2. Nat Genet 37: 166‐170, 2005.
 133. Mikami K , Strott CA . Cyclic AMP‐dependent protein kinase activity and protein phosphorylation in zones of the adrenal cortex. Biochem Biophys Res Commun 138: 895‐901, 1986.
 134. Miller WL , Strauss JF, 3rd . Molecular pathology and mechanism of action of the steroidogenic acute regulatory protein, StAR. J Steroid Biochem Mol Biol 69: 131‐141, 1999.
 135. Mohawk JA , Takahashi JS . Cell autonomy and synchrony of suprachiasmatic nucleus circadian oscillators. Trends Neurosci 34: 349‐358, 2011.
 136. Moore RY , Lenn NJ . A retinohypothalamic projection in the rat. J Comp Neurol 146: 1‐14, 1972.
 137. Mountjoy KG , Mortrud MT , Low MJ , Simerly RB , Cone RD . Localization of the melanocortin‐4 receptor (MC4‐R) in neuroendocrine and autonomic control circuits in the brain. Mol Endocrinol 8: 1298‐1308, 1994.
 138. Munck A , Guyre PM , Holbrook NJ . Physiological functions of glucocorticoids in stress and their relation to pharmacological actions. Endoc Rev 5: 25‐44, 1984.
 139. Nagoshi E , Saini C , Bauer C , Laroche T , Naef F , Schibler U . Circadian gene expression in individual fibroblasts: Cell‐autonomous and self‐sustained oscillators pass time to daughter cells. Cell 119: 693‐705, 2004.
 140. Nishi M , Kawata M . Brain corticosteroid receptor dynamics and trafficking: Implications from live cell imaging. Neuroscientist 12: 119‐133, 2006.
 141. Orchinik M , Murray TF , Franklin PH , Moore FL . Guanyl nucleotides modulate binding to steroid receptors in neuronal membranes. Proc Natl Acad Sci U S A 89: 3830‐3834, 1992.
 142. Orchinik M , Murray TF , Moore FL . A corticosteroid receptor in neuronal membranes. Science 252: 1848‐1851, 1991.
 143. Oster H , Damerow S , Hut RA , Eichele G . Transcriptional profiling in the adrenal gland reveals circadian regulation of hormone biosynthesis genes and nucleosome assembly genes. J Biol Rhythms 21: 350‐361, 2006.
 144. Oster H , Damerow S , Kiessling S , Jakubcakova V , Abraham D , Tian J , Hoffmann MW , Eichele G . The circadian rhythm of glucocorticoids is regulated by a gating mechanism residing in the adrenal cortical clock. Cell Metab 4: 163‐173, 2006.
 145. Park SY , Walker JJ , Johnson NW , Zhao Z , Lightman SL , Spiga F . Constant light disrupts the circadian rhythm of steroidogenic proteins in the rat adrenal gland. Mol Cell Endocrinol 371: 114‐123, 2013.
 146. Pfaff DW , Silva MT , Weiss JM . Telemetered recording of hormone effects on hippocampal neurons. Science 172: 394‐395, 1971.
 147. Picard D . Intracellular dynamics of the Hsp90 co‐chaperone p23 is dictated by Hsp90. Exp Cell Res 312: 198‐204, 2006.
 148. Pon LA , Hartigan JA , Orme‐Johnson NR . Acute ACTH regulation of adrenal corticosteroid biosynthesis. Rapid accumulation of a phosphoprotein. J Biol Chem 261: 13309‐13316, 1986.
 149. Pratt WB , Galigniana MD , Harrell JM , DeFranco DB . Role of hsp90 and the hsp90‐binding immunophilins in signalling protein movement. Cell Signal 16: 857‐872, 2004.
 150. Pratt WB , Morishima Y , Osawa Y . The Hsp90 chaperone machinery regulates signaling by modulating ligand binding clefts. J Biol Chem 283: 22885‐22889, 2008.
 151. Qian X , Droste SK , Gutierrez‐Mecinas M , Collins A , Kersante F , Reul JM , Linthorst AC . A rapid release of corticosteroid‐binding globulin from the liver restrains the glucocorticoid hormone response to acute stress. Endocrinology 152: 3738‐3748, 2011.
 152. Qian X , Droste SK , Lightman SL , Reul JM , Linthorst AC . Circadian and ultradian rhythms of free glucocorticoid hormone are highly synchronized between the blood, the subcutaneous tissue, and the brain. Endocrinology 153: 4346‐4353, 2012.
 153. Ralph MR , Foster RG , Davis FC , Menaker M . Transplanted suprachiasmatic nucleus determines circadian period. Science 247: 975‐978, 1990.
 154. Rankin J , Walker JJ , Windle R , Lightman SL , Terry JR . Characterizing dynamic interactions between ultradian glucocorticoid rhythmicity and acute stress using the phase response curve. PloS one 7: e30978, 2012.
 155. Reppert SM , Weaver DR . Coordination of circadian timing in mammals. Nature 418: 935‐941, 2002.
 156. Reul JM , de Kloet ER . Two receptor systems for corticosterone in rat brain: Microdistribution and differential occupation. Endocrinology 117: 2505‐2511, 1985.
 157. Reul JM , de Kloet ER . Anatomical resolution of two types of corticosterone receptor sites in rat brain with in vitro autoradiography and computerized image analysis. J Steroid Biochem 24: 269‐272, 1986.
 158. Reul JM , de Kloet ER , van Sluijs FJ , Rijnberk A , Rothuizen J . Binding characteristics of mineralocorticoid and glucocorticoid receptors in dog brain and pituitary. Endocrinology 127: 907‐915, 1990.
 159. Rivier C , Vale W . Interaction of corticotropin‐releasing factor and arginine vasopressin on adrenocorticotropin secretion in vivo. Endocrinology 113: 939‐942, 1983.
 160. Rose JD , Moore FL , Orchinik M . Rapid neurophysiological effects of corticosterone on medullary neurons: Relationship to stress‐induced suppression of courtship clasping in an amphibian. Neuroendocrinology 57: 815‐824, 1993.
 161. Rotsztejn W , Lalonde J , Normand M , Fortier C . Feedback inhibition of adrenocorticotropin release by corticosterone infusions in the adrenalectomized rat. Can J Physiol Pharmacol 53: 475‐478, 1975.
 162. Rotsztejn WH , Normand M , Lalonde J , Fortier C . Relationship between ACTH release and corticosterone binding by the receptor sites of the adenohypophysis and dorsal hippocampus following infusion of corticosterone at a constant rate in the adrenalectomized rat. Endocrinology 97: 223‐230, 1975.
 163. Russell GM , Henley DE , Leendertz J , Douthwaite JA , Wood SA , Stevens A , Woltersdorf WW , Peeters BW , Ruigt GS , White A , Veldhuis JD , Lightman SL . Rapid glucocorticoid receptor‐mediated inhibition of hypothalamic‐pituitary‐adrenal ultradian activity in healthy males. J Neurosci 30: 6106‐6115, 2010.
 164. Saphier D , Feldman S . Iontophoretic application of glucocorticoids inhibits identified neurones in the rat paraventricular nucleus. Brain Res 453: 183‐190, 1988.
 165. Sarabdjitsingh RA , Conway‐Campbell BL , Leggett JD , Waite EJ , Meijer OC , de Kloet ER , Lightman SL . Stress responsiveness varies over the ultradian glucocorticoid cycle in a brain‐region‐specific manner. Endocrinology 151: 5369‐5379, 2010.
 166. Sarabdjitsingh RA , Isenia S , Polman A , Mijalkovic J , Lachize S , Datson N , de Kloet ER , Meijer OC . Disrupted corticosterone pulsatile patterns attenuate responsiveness to glucocorticoid signaling in rat brain. Endocrinology 151: 1177‐1186, 2010.
 167. Sawchenko PE , Swanson LW , Vale WW . Co‐expression of corticotropin‐releasing factor and vasopressin immunoreactivity in parvocellular neurosecretory neurons of the adrenalectomized rat. Proc Natl Acad Sci U S A 81: 1883‐1887, 1984.
 168. Schoneveld OJ , Gaemers IC , Lamers WH . Mechanisms of glucocorticoid signalling. Biochim Biophys Acta 1680: 114‐128, 2004.
 169. Seale JV , Wood SA , Atkinson HC , Bate E , Lightman SL , Ingram CD , Jessop DS , Harbuz MS . Gonadectomy reverses the sexually diergic patterns of circadian and stress‐induced hypothalamic‐pituitary‐adrenal axis activity in male and female rats. J Neuroendocrinol 16: 516‐524, 2004.
 170. Seale JV , Wood SA , Atkinson HC , Harbuz MS , Lightman SL . Gonadal steroid replacement reverses gonadectomy‐induced changes in the corticosterone pulse profile and stress‐induced hypothalamic‐pituitary‐adrenal axis activity of male and female rats. J Neuroendocrinol 16: 989‐998, 2004.
 171. Seale JV , Wood SA , Atkinson HC , Harbuz MS , Lightman SL . Postnatal masculinization alters the HPA axis phenotype in the adult female rat. J Physiol 563: 265‐274, 2005.
 172. Seale JV , Wood SA , Atkinson HC , Lightman SL , Harbuz MS . Organizational role for testosterone and estrogen on adult hypothalamic‐pituitary‐adrenal axis activity in the male rat. Endocrinology 146: 1973‐1982, 2005.
 173. Shanks N , Lightman SL . The maternal‐neonatal neuro‐immune interface: Are there long‐term implications for inflammatory or stress‐related disease? J Clin Invest 108: 1567‐1573, 2001.
 174. Shanks N , Windle RJ , Perks PA , Harbuz MS , Jessop DS , Ingram CD , Lightman SL . Early‐life exposure to endotoxin alters hypothalamic‐pituitary‐adrenal function and predisposition to inflammation. Proc Natl Acad Sci U S A 97: 5645‐5650, 2000.
 175. Son GH , Chung S , Choe HK , Kim HD , Baik SM , Lee H , Lee HW , Choi S , Sun W , Kim H , Cho S , Lee KH , Kim K . Adrenal peripheral clock controls the autonomous circadian rhythm of glucocorticoid by causing rhythmic steroid production. Proc Natl Acad Sci U S A 105: 20970‐20975, 2008.
 176. Song KH , Park JI , Lee MO , Soh J , Lee K , Choi HS . LH induces orphan nuclear receptor Nur77 gene expression in testicular Leydig cells. Endocrinology 142: 5116‐5123, 2001.
 177. Spencer RL , Miller AH , Moday H , Stein M , McEwen BS . Diurnal differences in basal and acute stress levels of type I and type II adrenal steroid receptor activation in neural and immune tissues. Endocrinology 133: 1941‐1950, 1993.
 178. Spiga F , Harrison LR , Wood SA , MacSweeney CP , Thomson FJ , Craighead M , Grassie M , Lightman SL . Effect of the glucocorticoid receptor antagonist Org 34850 on fast and delayed feedback of corticosterone release. J Endocrinol 196: 323‐330, 2008.
 179. Spiga F , Liu Y , Aguilera G , Lightman SL . Temporal effect of adrenocorticotrophic hormone on adrenal glucocorticoid steroidogenesis: Involvement of the transducer of regulated cyclic AMP‐response element‐binding protein activity. J Neuroendocrinol 23: 136‐142, 2011.
 180. Spiga F , Waite EJ , Liu Y , Kershaw YM , Aguilera G , Lightman SL . ACTH‐Dependent Ultradian Rhythm of Corticosterone Secretion. Endocrinology 152: 1448‐1457, 2011.
 181. Stavreva DA , Wiench M , John S , Conway‐Campbell BL , McKenna MA , Pooley JR , Johnson TA , Voss TC , Lightman SL , Hager GL . Ultradian hormone stimulation induces glucocorticoid receptor‐mediated pulses of gene transcription. Nat Cell Biol 11: 1093‐1102, 2009.
 182. Stephan FK , Zucker I . Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc Natl Acad Sci U S A 69: 1583‐1586, 1972.
 183. Sternberg EM , Hill JM , Chrousos GP , Kamilaris T , Listwak SJ , Gold PW , Wilder RL . Inflammatory mediator‐induced hypothalamic‐pituitary‐adrenal axis activation is defective in streptococcal cell wall arthritis‐susceptible Lewis rats. Proc Natl Acad Sci U S A 86: 2374‐2378, 1989.
 184. Sternberg EM , Young WS, 3rd , Bernardini R , Calogero AE , Chrousos GP , Gold PW , Wilder RL . A central nervous system defect in biosynthesis of corticotropin‐releasing hormone is associated with susceptibility to streptococcal cell wall‐induced arthritis in Lewis rats. Proc Natl Acad Sci U S A 86: 4771‐4775, 1989.
 185. Stocco DM , Clark BJ . Regulation of the acute production of steroids in steroidogenic cells. Endoc Rev 17: 221‐244, 1996a.
 186. Stocco DM , Clark BJ . Role of the steroidogenic acute regulatory protein (StAR) in steroidogenesis. Biochem Pharmacol 51: 197‐205, 1996b.
 187. Stokkan KA , Yamazaki S , Tei H , Sakaki Y , Menaker M . Entrainment of the circadian clock in the liver by feeding. Science 291: 490‐493, 2001.
 188. Stratmann M , Schibler U . Properties, entrainment, and physiological functions of mammalian peripheral oscillators. J Biol Rhythms 21: 494‐506, 2006.
 189. Sugawara T , Holt JA , Kiriakidou M , Strauss JF, 3rd . Steroidogenic factor 1‐dependent promoter activity of the human steroidogenic acute regulatory protein (StAR) gene. Biochemistry 35: 9052‐9059, 1996.
 190. Takemori H , Kanematsu M , Kajimura J , Hatano O , Katoh Y , Lin XZ , Min L , Yamazaki T , Doi J , Okamoto M . Dephosphorylation of TORC initiates expression of the StAR gene. Mol Cell Endocrinol 265‐266: 196‐204, 2007.
 191. Takemori H , Okamoto M . Regulation of CREB‐mediated gene expression by salt inducible kinase. J Steroid Biochem Mol Biol 108: 287‐291, 2008.
 192. Tapp WN , Holaday JW , Natelson BH . Ultradian glucocorticoid rhythms in monkeys and rats continue during stress. Am J Physiol 247: R866‐R871, 1984.
 193. Tasker JG , Herman JP . Mechanisms of rapid glucocorticoid feedback inhibition of the hypothalamic‐pituitary‐adrenal axis. Stress 14: 398‐406, 2011.
 194. Tousson E , Meissl H . Suprachiasmatic nuclei grafts restore the circadian rhythm in the paraventricular nucleus of the hypothalamus. J Neurosci 24: 2983‐2988, 2004.
 195. Ulrich‐Lai YM , Arnhold MM , Engeland WC . Adrenal splanchnic innervation contributes to the diurnal rhythm of plasma corticosterone in rats by modulating adrenal sensitivity to ACTH. Am J Physiol Regul Integr Comp Physiol 290: R1128‐R1135, 2006.
 196. Ulrich‐Lai YM , Herman JP . Neural regulation of endocrine and autonomic stress responses. Nat Rev Neurosci 10: 397‐409, 2009.
 197. Vale W , Spiess J , Rivier C , Rivier J . Characterization of a 41‐residue ovine hypothalamic peptide that stimulates secretion of corticotropin and beta‐endorphin. Science 213: 1394‐1397, 1981.
 198. Van Eekelen JA , De Kloet ER . Co‐localization of brain corticosteroid receptors in the rat hippocampus. Prog Histochem Cytochem 26: 250‐258, 1992.
 199. Vrang N , Larsen PJ , Mikkelsen JD . Direct projection from the suprachiasmatic nucleus to hypophysiotrophic corticotropin‐releasing factor immunoreactive cells in the paraventricular nucleus of the hypothalamus demonstrated by means of Phaseolus vulgaris‐leucoagglutinin tract tracing. Brain Res 684: 61‐69, 1995.
 200. Waite EJ , McKenna M , Kershaw Y , Walker JJ , Cho K , Piggins HD , Lightman SL . Ultradian corticosterone secretion is maintained in the absence of circadian cues. Eur J Neurosci 36: 3142‐3150, 2012.
 201. Walker JJ , Spiga F , Waite E , Zhao Z , Kershaw Y , Terry JR , Lightman SL . The origin of glucocorticoid hormone oscillations. PLoS Biol 10: e1001341, 2012.
 202. Walker JJ , Terry JR , Lightman SL . Origin of ultradian pulsatility in the hypothalamic‐pituitary‐adrenal axis. Proc Biol Sci R Soc 277: 1627‐1633, 2010.
 203. Wallberg AE , Flinn EM , Gustafsson JA , Wright AP . Recruitment of chromatin remodelling factors during gene activation via the glucocorticoid receptor N‐terminal domain. Biochem Soc Trans 28: 410‐414, 2000.
 204. Watts AG , Swanson LW . Efferent projections of the suprachiasmatic nucleus: II. Studies using retrograde transport of fluorescent dyes and simultaneous peptide immunohistochemistry in the rat. J Comp Neurol 258: 230‐252, 1987.
 205. Watts AG , Swanson LW , Sanchez‐Watts G . Efferent projections of the suprachiasmatic nucleus: I. Studies using anterograde transport of Phaseolus vulgaris leucoagglutinin in the rat. J Comp Neurol 258: 204‐229, 1987.
 206. Watts AG , Tanimura S , Sanchez‐Watts G . Corticotropin‐releasing hormone and arginine vasopressin gene transcription in the hypothalamic paraventricular nucleus of unstressed rats: Daily rhythms and their interactions with corticosterone. Endocrinology 145: 529‐540, 2004.
 207. Weitzman ED , Fukushima D , Nogeire C , Roffwarg H , Gallagher TF , Hellman L . Twenty‐four hour pattern of the episodic secretion of cortisol in normal subjects. J Clin Endocrinol Metab 33: 14‐22, 1971.
 208. Welsh DK , Yoo SH , Liu AC , Takahashi JS , Kay SA . Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression. Curr Biol 14: 2289‐2295, 2004.
 209. Widmaier EP , Dallman MF . The effects of corticotropin‐releasing factor on adrenocorticotropin secretion from perifused pituitaries in vitro: Rapid inhibition by glucocorticoids. Endocrinology 115: 2368‐2374, 1984.
 210. Wilkinson CW , Shinsako J , Dallman MF . Return of pituitary‐adrenal function after adrenal enucleation or transplantation: Diurnal rhythms and responses to ether. Endocrinology 109: 162‐169, 1981.
 211. Windle RJ , Wood SA , Kershaw YM , Lightman SL , Ingram CD . Adaptive changes in basal and stress‐induced HPA activity in lactating and post‐lactating female rats. Endocrinology 154: 749‐761, 2013.
 212. Windle RJ , Wood SA , Kershaw YM , Lightman SL , Ingram CD , Harbuz MS . Increased corticosterone pulse frequency during adjuvant‐induced arthritis and its relationship to alterations in stress responsiveness. J Neuroendocrinol 13: 905‐911, 2001.
 213. Windle RJ , Wood SA , Lightman SL , Ingram CD . The pulsatile characteristics of hypothalamo‐pituitary‐adrenal activity in female Lewis and Fischer 344 rats and its relationship to differential stress responses. Endocrinology 139: 4044‐4052, 1998.
 214. Windle RJ , Wood SA , Shanks N , Lightman SL , Ingram CD . Ultradian rhythm of basal corticosterone release in the female rat: Dynamic interaction with the response to acute stress. Endocrinology 139: 443‐450, 1998.
 215. Wochnik GM , Ruegg J , Abel GA , Schmidt U , Holsboer F , Rein T . FK506‐binding proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J Biol Chem 280: 4609‐4616, 2005.
 216. Yamazaki S , Numano R , Abe M , Hida A , Takahashi R , Ueda M , Block GD , Sakaki Y , Menaker M , Tei H . Resetting central and peripheral circadian oscillators in transgenic rats. Science 288: 682‐685, 2000.
 217. Zazopoulos E , Lalli E , Stocco DM , Sassone‐Corsi P . DNA binding and transcriptional repression by DAX‐1 blocks steroidogenesis. Nature 390: 311‐315, 1997.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Francesca Spiga, Jamie J. Walker, John R. Terry, Stafford L. Lightman. HPA Axis‐Rhythms. Compr Physiol 2014, 4: 1273-1298. doi: 10.1002/cphy.c140003