Sei sulla pagina 1di 16

Hypothalamic-Pituitary-Adrenal (HPA) Axis

and Aging
Deepashree Gupta1 and John E. Morley*2

ABSTRACT
Human aging is associated with increasing frailty and morbidity which can result in signifi-
cant disability. Dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis may contribute to
aging-related diseases like depression, cognitive deficits, and Alzheimer’s disease in some older
individuals. In addition to neuro-cognitive dysfunction, it has also been associated with declin-
ing physical performance possibly due to sarcopenia. This article reviews the pathophysiology
of HPA dysfunction with respect to increased basal adrenocorticotropic hormone (ACTH) and
cortisol secretion, decreased glucocorticoid (GC) negative feedback at the level of the paraven-
tricular nucleus (PVN) of the hypothalamus, hippocampus (HC), and prefrontal cortex (PFC), and
flattening of diurnal pattern of cortisol release. It is possible that the increased cortisol secretion
is secondary to peripheral conversion from cortisone. There is a decline in pregnolone secretion
and C-19 steroids (DHEA) with aging. There is a small decrease in aldosterone with aging, but
a subset of the older population have a genetic predisposition to develop hyperaldosteronism
due to the increased ACTH stimulation. The understanding of the HPA axis and aging remains a
complex area with conflicting studies leading to controversial interpretations.  C 2014 American

Physiological Society. Compr Physiol 4:1495-1510, 2014.

Introduction Human aging is known to be associated with progressive


frailty and morbidity. The World Health Organization (WHO)
The release of corticotropin-releasing hormone (CRH), has estimated that in our lifetime, the number of elderly (ages
adrenocorticotropic hormone (ACTH), and cortisol in 65 and up) will triple with a significant reduction in the pro-
response to stress is a vital adaptive response of the body. portion of children. With increasing human longevity, there
Stimulation of the hypothalamic-pituitary-adrenal (HPA) axis is an increasing focus on research to prevent or delay dis-
not only provides a quick physical response to stress but also abilities associated with aging (67, 125). The HPA hormonal
mediates long-term changes in memory centers in the brain axis has an important role in maintaining homeostasis and
leading to formation of long-term memories of the experience. dysfunction of this axis is one of several plausible pathways
This is an important adaptive mechanism for the body to effec- contributing to biological aging (80). Abnormal GC secre-
tively counteract similar stressors in the future. However, if tion is thought to be involved in many aging-related diseases,
this stress reaction is enhanced and/or persistent long term, it including depression, cognitive deficits, and Alzheimer’s dis-
can lead to damage to certain areas of the brain specifically the ease (55, 143, 147, 215). Thus, understanding and modifying
prefrontal cortex (PFC), paraventricular neurons (PVN), and factors underlying abnormal GC signaling may be of consid-
hippocampus (HC) which are important areas for cognition erable importance in the prevention and treatment of aging-
and memory formation. These areas are also important for related diseases (163).
mediating negative cortisol feedback on HPA axis, and dam- ACTH, the principal hormone stimulating adrenal GC
age to these areas perpetuates a vicious cycle of glucocorticoid biosynthesis and secretion, is synthesized in the anterior pitu-
excess. Also the absolute number of GC receptors (GR) and itary as part of a much larger precursor molecule called
mineralocorticoid or type I receptors (MRs) in these areas pro-opiomelanocortin (POMC) (Fig. 1). POMC gets cleaved
decreases with aging, further blunting the cortisol mediated into β-lipoprotein and pro-ACTH in the anterior pituitary
negative feedback mechanism. Decrease in numbers of MRs
in the HC are thought to increase basal ACTH and cortisol
concentrations during the quiescent phase-nadir of the circa- * Correspondence to morley@slu.edu
dian rhythm, when circulating cortisol predominantly binds 1 Division of Endocrinology, Saint Louis University, St. Louis, Missouri
to MRs. This article discusses these pathologic changes in 2 Divisions of Endocrinology and Geriatric Medicine, Saint Louis
detail and the lab abnormalities that accompany them. There University, St. Louis, Missouri
is increased interest in research is this exciting field with Published online, October 2014 (comprehensivephysiology.com)
respect to diet (135, 192) and lifestyle changes which can DOI: 10.1002/cphy.c130049
prevent or reverse morbidity that accompanies aging. Copyright  C American Physiological Society.

Volume 4, October 2014 1495


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

Glucocorticoids (GCs) in brain


MR/GR
− (HC) Mineralocorticoid • Fast/membrane Glucocorticoid
PVN (type I) receptor actions (type II) receptor
− • Slow/genomic
actions
CRH • Mainly fou and in limbic • Ubiquitous
− region (hippocampus) distribution in brain
• High affinity for GCs • Low affinity for GCs
+ • Feedback inhibition at
nadir of circadian
• Feedback inhibition
after stress response
Anterior pituitary rhythm • Mediate termination
Posterior pituitary • Mediate initiation of of stress response
stress response • Promote memory
AVP Stressors • Promote appraisal of a consolidation by

+ IL‐1 novel condition, processing and storing


behavioral flexibility, and experience
POMC IL‐1 selective attention

+ TNF ‐ α
ACTH
− Figure 2 Glucocorticoid receptors.

Adrenal
via intracellular nuclear receptors termed glucocorticoid or
type II receptor (GR) and mineralocorticoid or type I receptor
(MR), respectively (Fig. 2); they consist of a carboxy-terminal
Glucocorticoids ligand-binding domain, a central DNA-binding domain and
an N-terminal hypervariable region. The genomic effects of
Figure 1 Normal HPA axis.
GCs on MRs and GRs are typically slow in nature; they can
occur in approximately 15 to 30 min after receptor activation
and the latter further gets cleaved into ACTH, joining pep- and may last from less than 1 h up to days depending upon
tide and amino-terminal peptide (58, 228). POMC secretion duration of exposure to GCs (59, 118, 170, 171).
is tightly controlled by factors such as CRH, arginine vaso- There recently has been a discovery of “fast/membrane”
pressin (AVP) (138, 186), endogenous circadian rhythm, mode of action of MRs and GRs in addition to the
stress, and lastly but importantly, feedback inhibition by cor- “slow/nuclear” action as described above. This is thought to
tisol. CRH is a 41-amino-acid peptide synthesized in the be mediated via a nongenomic pathway involving membrane-
PVN of the hypothalamus (4, 238). It is then secreted into the located MRs and GRs leading to GC-dependent enhancement
hypophyseal portal blood and binds to type 1 CRH receptors of glutamate in CA1 hippocampal neurons; these membrane
on anterior pituitary corticotrophs to stimulate POMC gene receptors have low ligand affinity compared to nuclear recep-
transcription (37). AVP potentiates CRH-mediated POMC tors (115, 122, 237). The nongenomic effects occur within
and hence ACTH secretion by acting on V1B receptor (97). minutes after GC application and have been reported to have
Pro-inflammatory cytokines like interleukin 1 (IL-1), IL-6, behavioral effects like novelty-induced locomotor activity and
and tumor necrosis factor (TNF)-α and physical stresses can aggressiveness in rats (160, 215).
also augment ACTH secretion, as a direct effect as well as cen- The human body responds to stressors through differ-
trally via CRH (11, 38). ACTH binds to G protein-coupled, ent physiologic responses. The first response is activation of
melanocortin -2 receptors (MC2R) on adrenocortical cells sympathetic nervous system and release of adrenaline and
(172) to exert both immediate and chronic effects on the noradrenaline from the adrenal medulla, called the “fight or
adrenal gland. Acutely adrenal steroidogenesis is increased flight” response (30, 78). Stressors also activate the HPA axis
via StAR-mediated cholesterol delivery to CYP11A1 enzyme by increasing ACTH and GC levels through central actions
in the inner mitochondrial membrane (233). Within 24 h of of CRH and AVP. Elevation of GCs as a counterregulatory
exposure, ACTH exerts effects at the transcriptional level mechanism to fever, hypoglycemia (69), hypotension, and
to increase synthesis of all steroidogenic CYP enzymes— exercise (137) is well documented in literature. GCs mobilize
CYP11A1, CYP17A1, CYP21A2 as well as CYP11B1 and energy by activating proteolysis and lipolysis and inhibiting
hence stimulates GC production (227, 252); it also increases glycolysis and immunogenesis in different tissues, to cope
adrenal weight by inducing both hyperplasia and hypertrophy. with energy demands triggered by the stressor and to restore
GCs in most mammals is cortisol and corticosterone (CORT) homeostasis (219). When homeostasis has been recovered,
in rodents; however, CORT can be found in human blood high levels of GCs are tightly regulated by a negative GC
as well (29). The effect of ACTH on aldosterone secretion feedback mechanism at the level of the pituitary, PVN, and
is modest; acutely aldosterone secretion could increase due HC and this modulates circadian activity of the HPA axis as
to ACTH induced stimulation of early pathways of adrenal well as its stress response (77, 101, 113, 151, 154, 215, 241).
steroidogenesis but this effect is lost with chronic ACTH stim- GCs play at least three functions in the brain. The first is,
ulation. Both GCs and aldosterone exert their genomic effects like in the rest of the body, to regulate energy availability

1496 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

during the stress response. The second is to promote behav- aging is thought to have an adverse impact on mnesic activi-
ioral adaptation and regulate (either enhance or impair) sev- ties (45). This can lead to an exaggerated or inadequate initial
eral memory processes (114). The third function of GCs in HPA stress response, impaired containment, delayed recov-
the brain is control of the HPA axis activity through GRs. ery, and compromised adaptation, which in turn can make
GCs inhibit POMC gene transcription in the anterior pitu- the brain susceptible to affective pathologies (47). In mice,
itary (138) and CRH and AVP mRNA synthesis and secretion global reduction in GR signaling activates the HPA axis and
in the hypothalamus (44, 124). However, GC feedback reg- impairs cognition (185,206), while deletion of GR in forebrain
ulation of hypothalamic CRH neurons and hippocampal and only increases corticosterone levels and increases depressive-
PFC pyramidal neurons can become active within seconds like behavior (24, 239). In contrast, forebrain-specific dele-
of application of a brief stressor, largely through modulation tion of MR leads to learning and memory impairment (17),
of excitatory or inhibitory synaptic inputs to these neurons while forebrain overexpression of MR enhances memory and
(108, 204). MRs are predominantly expressed in the limbic reduces anxiety (134, 211).
structures in the brain, mainly the HC and some are also
found in the hypothalamus (75, 105). GR distribution is more
ubiquitous in the brain and highest density is found in the
Glucocorticoid Cascade Hypothesis
parvocellular PVN (hypothalamus), in neurons of ascending
aminergic pathways and in limbic neurons controlling PVN Stress is an important adaptive response of the body, however
activation trans-synaptically. Coexpression of MR and GR is if this response is maintained for more time than necessary
most abundant in hippocampal pyramidal cells and amygdale or if chronically activated, harmful effects on the body like
in almost all species (45, 48, 226). immunosuppression, peripheral muscle dysfunction, and neu-
In the brain, especially in the HC, 11 β-hydroxysteroid ronal death can appear; this condition is also referred to as
dehydrogenase type 1 (11 β-HSD-1) which regenerates active allostatic load (150). Hence stress has been studied as one
glucocorticoids, is widely expressed but the type 2 isoenzyme of the factors contributing to unsuccessful aging (152). In
of this enzyme, which rapidly inactivates glucocorticoids is 1986, Sapolsky et al. (218), proposed the “Glucocorticoid
undetectable (47, 76, 113). As a result, MRs bind GCs with a cascade hypothesis” which suggests that GCs secreted during
10-fold higher affinity than GRs and lose their MR selectivity times of stress desensitize the HC to further GC exposure
(47, 203). Binding of GCs to high affinity mineralocorticoid by downregulating GRs. Initially this effect is self-correcting
receptors plays an important role in negative feedback control and hence reversible, however, long-standing downregulation
and maintenance of basal HPA axis mainly during the nadir of GRs and impairment in the negative GC feedback mecha-
of the circadian rhythm (42, 47) while binding of GCs to low nism in aged rats leads to an enhanced exposure of the brain
affinity glucocorticoid receptors plays an important role in to the deleterious effects of GCs which, in turn, would perma-
feedback control of HPA axis during stress (180). Chronic nently reduce the number of GC-sensitive neurons in the HC
HPA axis inhibition by tonic elevation of plasma GC levels (site of learning and memory) and consequently lead to age-
can regain responsiveness to new acutely applied stressors related cognitive deficits. The damage to HC would further
by inhibiting noradrenergic neurons via CRH neurons in the reduce the expression of GRs and MRs leading to decreased
PVN (81). accuracy of the inhibitory control of GCs, which in turn would
Specific areas of the brain are activated during the time further potentiate damage to the HC, also partly due to gradual
of stress-amygdala for emotional response and formation of increase in GCs over time (20, 22, 191, 224, 257). The conclu-
emotional memories (153,193), the HC for memory and learn- sion about neuronal death caused by GCs and stress remains
ing processes (214), and the frontal cortex for planning and controversial. In young rodents, while chronic injection of
controlling actions. Together they play an important role in exogenous stress levels of corticosterone produced neuronal
managing emotions, neuroendocrine responses, and cognitive loss of CA3 comparable to aged rats (217), this effect was
learning (199). MRs play an important role in the stress cas- observed only if treatment began when the rats were juvenile
cade and when activated, amplify the initial stress event (117) and not if prolonged corticosterone exposure occurred only
which is followed by MR mediated neuronal stability and during adulthood (230). Recent studies by Sapolsky (216-218)
integrity. GR-mediated feedback contains the stress process suggest that excessive glucocorticoid stress response may
and GRs promote memory consolidation by processing and actually augment aspects of inflammation during acute brain
storing the experience (171, 183, 185, 203, 220). Brain MRs injury. Two acute neurological injury rat models—Kainic acid
are also thought to be involved in appraisal of a novel con- (KA)-induced excite toxic injury to HC and middle cerebral
dition, behavioral flexibility (17,184,188), selective attention artery occlusion (MCAO) model of stroke were studied and
and emotions like anxiety (25). Hence MR and GR operate in findings supported that GCs can act directly activate myeloid
balance and mediate initiation (possibly via membrane MR) cells. Myeloid cells migrate to the injury site and in this study
(122) and termination of the HPA stress response; termination increased nuclear p65 levels and decreased anti-inflammatory
is thought to be mediated via negative feedback at intracel- signaling of CX3CL1 after KA administration and produced
lular GR, but also MR (93). Compromise of this MR/GR more IL-6 in MCAO model; this is contrary to the classi-
interaction, particularly in the limbic system neurons during cal expectation of GCs suppressing immune cell activation

Volume 4, October 2014 1497


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

(229). This could partially be explained by differing action of Anatomy Hormones and receptors
GCs on different brain areas. The classic anti-inflammatory
effects of GCs are typically seen in subcortical regions like
the substantia nigra (210) and hypothalamus (175), whereas
Paraventricular Glucocorticoid
their immune-activating properties have been primarily seen
nucleus receptor
in forebrain regions like the hippocampus and frontal cortex
(49, 176). In these forebrain regions, even chronic exposure
to GCs (1-2 weeks) can be immune activating, especially if
Portal vein CRF Peripheral
it occurs before the injury (175). In addition, GC exposure organ
before an injury has been shown to augment aspects of the response
subsequent inflammatory response to several types of CNS
injury (49,52,72,73,144,175,176). The benefits of exogenous Pituitary
steroid administration in decreasing vasogenic edema sur-
rounding brain tumors, abscesses, after neurosurgical manip-
ulation and in some inflammatory and auto-immune diseases Cortisol
like SLE are well known. However, steroids are relatively Blood
ACTH
ineffective for cytotoxic and vasogenic brain edema during
and after ischemic injury (112) and are contraindicated for
reducing intra cranial pressure in patients with severe Trau-
matic Brain Injury after the multicenter CRASH trial revealed Adrenal
increased all-cause mortality in patients who received corti-
costeroids vs. the placebo group (207).
Figure 3 Effects of aging on the rodent hypothalamic-pituitary-
Plasma GC gets suppressed by dexamethasone (DEX) adrenal axis.
infusions into each brain region of young rats implying that
each region has a role as the feedback site in the brain (164).
DEX is a synthetic GC that inhibits HPA axis via negative basal ACTH and cortisol concentrations during the quiescent
feedback and subsequently decreases release of endogenous phase-nadir of the circadian rhythm, when circulating corti-
cortisol (71, 234). However, when DEX was infused into sol predominantly binds to the MRs. (50, 57, 82, 244). The
each brain region of aged rats, no suppressive response was MR:GR balance hypothesis states that MR and GR imbal-
observed, indicating that GC negative feedback ability at the ance can lead to neuroendocrine dysregulation and impaired
brain level is markedly decreased in aged rats. Also in aged behavioral adaptation, potentially aggravating aging-related
rats, the GC negative feedback was enhanced at the systemic neurocognitive deterioration (183). Downregulation of hip-
levels but attenuated oppositely at the brain level (163). pocampal MRs in aged animals, rodents, dogs as well as
In the 1980s and 1990s, several studies showed that lesions primates, has been a consistent finding (46, 82, 106, 204), and
of the HC lead to an enhanced production of GCs under acute strategies to enhance MR expression were associated with
stress conditions (104,114,121,219) thus suggesting that neg- enhanced cognitive performance. The gain-of-function muta-
ative GC feedback at the level of HC plays a major inhibitory tion in the MR in the Brown Norway rat strain was shown to
role on HPA axis activity (20, 79, 80). In dogs, it was shown confer successful aging (148). Also starting tricyclic antide-
that hippocampal volume as well as the number of hippocam- pressants in midlife and continuing them until senescence
pal mineralocorticoid receptors decrease with age (113). Stud- ameliorated cognitive impairment in aged rats, thought to
ies in rodents suggest that the hypothalamic pituitary adrenal be due to upregulation of MR paralleled by lower corticos-
axis with aging is overactive (Fig. 3). There is a reduction in terone secretion (261). The HC is crucial in memory stor-
both type I (mineralocorticoid) and type II (glucocorticoid) age and retrieval and plays an important role in declarative
receptors in the hippocampus resulting in a decreased inhibi- memory. The effect of GCs on brain functions, in particu-
tion of PVN in the hypothalamus (123, 135, 178). This results lar, hippocampus related memory performance is now well
in an attenuation of glucocorticoid feedback resulting in an documented. It has been described as showing an inverted
increase in portal venous CRH and circulating ACTH, deoxy- U-shape pattern, with too low or too high levels being associ-
corticosterone, and corticosterone (18, 27, 29, 43, 96, 98). As ated with impairment while moderate levels enhance perfor-
GC and ACTH levels increase with aging (156), they sup- mance (61,87,120,139,141,159,182,218,223,233,234,258).
press basal AVP levels by feedback inhibition and hence Hypercortisolism has been associated with reduced cell sur-
basal portal AVP concentrations were found to be signifi- vival, proliferation, and neurogenesis in the brain especially
cantly lower in aged compared to young rats. When aged at the limbic levels of the hippocampal subfield, CA1, CA3,
rats underwent bilateral adrenalectomy followed by physio- and dentate gyrus cells (116). Expression levels and signals
logic GC replacement, basal portal AVP levels normalized to through GRs as well as MRs are likely to decrease in the aged
that observed in young intact animals (173). Age associated hippocampus. Aged rats exhibiting impaired spatial mem-
decrease in hippocampal MRs is hypothesized to increase ory show decreases in GR density and GR mRNA in the

1498 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

hippocampus (18, 111). Also, nuclear transport and subse- Disability


quent binding of GRs to the consensus sequence on genes,
that is, GC responsive element, are decreased in the aged Frailty Depression CRF Anorexia
Weight
loss
hippocampus, suggesting the reduced ability of GRs to trans-
duce GC signals (178). The PFC also has an important role Sarcopenia Infections
in maintaining cognitive and memory functions; these are
frequently impaired in elderly subjects. Dysregulation of the Muscle
Immune
function
HPA axis has been shown to be associated with smaller vol- wasting
ume of medial PFC in elderly humans (143). Cortisol Neuronal damage
To some extent, this HPA overactivity is offset by a
decrease in CBG (Cortisol Binding Globulin), resulting in Cognition
Osteopenia
an increase in free corticosterone in older rats (155). The
“glucocorticoid cascade hypothesis” has not been consistently Visceral Insulin Mild
Hip fracture Hypertension cognitive
reproduced in subsequent studies using rats and other models. obesity resistance
impairment
In aged Brown Norway rats, corticosterone levels were very
low but they demonstrated healthy aging (245, 246). Some
Atherosclerosis
of these contradictory findings could be from species-specific Dementia
differences in stress steroid receptor expression (MR and GR
distribution in HC as discussed above). While GRs and MRs Coronary artery
were quite concentrated in the HC of rodent brain (215), disease
non-human primates like Rhesus monkeys express relatively
fewer GRs in the HC but abundant GRs in the hypothalamus Figure 4 Effects of elevated cortisol levels that can accelerate the
aging process.
and pituitary (213). These conflicting findings question the
role of GRs in mediating neuronal death in the HC. Stress
responses in older rats are variable, in some cases showing an stressors, may have dementia or depression, altered sleep pat-
increase and in others no change (103). The ACTH response terns including sleep apnea, obesity or weight loss, alcohol
to hemorrhage in older rats was only 50% of that seen in use and use multiple medications. All of these factors can alter
younger rats (98). Aging Peromyscus californicus showed the HPA axis. In addition, gender differences related to sex
minimal changes in the hypothalamic-pituitary-adrenal axis steroid changes, have not been well studied in older persons.
(94). There is a decrease in portal AVP with aging which For these reasons, the human literature has shown a variety of
as mentioned above, stimulates ACTH production from the contradictory effects.
pituitary in concert with CRH (84). In aged rodents, there is In a prospective study of aging humans, subjects with ris-
a decline in GR number, but not affinity, in peripheral tissues ing cortisol levels during the latest four years demonstrated
(53). This is accompanied by alterations in the glucocorticoid impaired explicit memory with a 14% reduction in hippocam-
inducible tyrosine amino transferase and tryptophan oxyge- pal volume (140). As discussed above, high-affinity MRs are
nase activities in rat liver; these could play a role in changes thought to be responsible for tonic inhibition of HPA activ-
of cell responses to GCs seen with aging (205). Studies in ity during the nadir of the circadian rhythm (15). Conflicting
aged monkeys found an increase in evening cortisol in aged results were elicited when human studies were performed
female rhesus monkeys, an increase in response to CRH and using MR antagonists, like spironolactone to study cortisol
a decrease in cortisol escape after DEX (90).

Table 1 Effects of Aging on the Hypothalamic Pituitary Glucocorti-


coid Axis in Humans based on the Best Judgment in Available Literature
Human Studies
Many aging changes are similar to those seen in persons with r Urinary free cortisol unchanged
r Increased 24 h total and free plasma cortisol
hypercortisolemia (Fig. 4). This has led to the concept that r Increased daily cortisol secretion based on salivary cortisol
the HPA axis is overactive with aging. However, the data to r Phase advancement of morning cortisol
support this is limited. The Glucocorticoid Cascade Hypoth- r Decreased amplitude of 24 h cortisol secretion
r Increased fragmentation of cortisol secretion
esis which utilized rats was not reproducible consistently in r No change in cortisol binding globulin
human beings and these inconsistencies drew into question the r Increased corticosterone production rate
relevance of Glucocorticoid Cascade Hypothesis for humans r Small decrease in plasma clearance rate of cortisol
r Increased post dexamethasone cortisol levels
(174). Table 1 summarizes the most likely effects of aging on r No change to ACTH response to CRH but a blunted ability of
the hypothalamic-pituitary-adrenal axis in humans. dexamethasone to suppress the response
Measuring the HPA axis in older persons is fraught with r Decreased cortisol response to ACTH
r Older persons had a lower ACTH and cortisol response to a
difficulty. Many older persons live under a variety of illnesses psychosocial stressor
which alter cytokine levels, have multiple psychological

Volume 4, October 2014 1499


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

secretion during the circadian rhythm; this could partially levels around midnight. In older persons, this circadian
be attributed to the different doses and experimental mod- rhythm is flattened (65, 249). In persons over 70 years of age,
els used for these studies. Some studies showed increased cortisol tends to increase in the evening, though not in all stud-
cortisol secretion during both peak and trough of the cir- ies (128, 161, 243, 244). The time of the cortisol nadir tends
cadian rhythm (51, 262), while others showed no discern- to be shorter. These changes are associated with a decrease
able effect of spironolactone on basal HPA activity or HPA in rapid eye movement sleep. The maximum levels of early
response to CRH stimulation (14, 53, 160). In normal young morning cortisol tend to occur earlier, that is, exhibiting phase
subjects, administration of a more selective MR antago- advancement (161,244). These changes lead to smaller ampli-
nist, canreonate, either as a short infusion or as prolonged tude in the circadian amplitude of cortisol secretion (243).
treatment in different doses, induced significant increase in There is an increase in 24 h plasma free and total cortisol
HPA activity during the nadir phase of the circadian rhythm levels in older men and women (198). This increase is corre-
(5,16,19,56,82,89,146). High doses of MR antagonists were lated with an increase in corticosteroid production rate. They
shown to enhance HPA response to both CRH and AVP stim- also found an increase in lunch-time food associated corti-
ulations suggesting increased CRH and AVP levels mediate sol with aging. There was no change in CBG. The data for
the stimulatory effect of MR blockade (5, 16, 106). Blocking plasma clearance rate (PCR) for cortisol and aging is less
of CNS MRs after pretreatment with canreonate was capa- clear. Older data suggested an increase in half-life with aging
ble of enhancing the ACTH and cortisol responses to CRH and a decrease in PCR (225, 240, 256). However, each of
during sleep, but not during wakefulness (21). Another study these studies included using a very high dose of tracer and
showed that subchronic treatment with canreonate amplified assumed a single-pool model. Barton et al. (9, 10) used mod-
cortisol secretion during physical activity and this remained ern methodology and found small, nonsignificant, decrease in
elevated during subsequent rest (255). In elderly humans, PCR in older men and women. Values in women tended to be
acute administration of canreonate showed increased HPA lower.
axis activity during the nadir of the circadian rhythm; this Conventional assessment of only total cortisol may under-
response, however, was less marked than younger subjects, estimate the actual extent of hypercortisolism in the elderly.
possibly due to age-related MR function impairment (82). Kudielka et al. (127) showed that there was an age-related
Subchronic treatment of aging individuals with spironolac- reduction in CBG in older men, though this was not seen
tone not only increased HPA axis activity during nadir of by others (191). Salivary cortisol is considered a reasonable
circadian rhythm, but also increased circadian peak cortisol proxy for free cortisol. Salivary cortisol levels showed a ten-
and salivary cortisol concentrations (106). dency to increase with age even when measured at different
Studies with MR agonists like fludrocortisone (FC) also times of the day suggestive of age-dependent increases in
provided interesting data on HPA axis activity. In young basal HPA activity (40, 179). Dmitrieva et al. (54) found that
healthy human subjects, high pharmacologic doses of FC can older males, especially, if they used medications, were more
significantly inhibit cortisol levels during nadir of circadian likely to have slightly higher salivary cortisol levels with a flat-
rhythm and reduce stimulatory effect of metyrapone on ACTH tened diurnal pattern. When an appropriate protocol to include
and 11-deoxycorticosterone (28,187,189). On the other hand, only healthy older persons was used, there was no difference
acute administration of FC showed only a minor reduction of in urinary free cortisol (UFC) in older persons (34). UFC was
HPA activity in elderly subjects (187). Hence based on cur- slightly lower in older women than men and this has been
rent evidence, there is an increased age associated liability confirmed in other studies (85, 236). There is a decline in
of human HPA axis particularly with respect to impairment urinary aldosterone (236). Higher UFC levels are present in
in central MRs; this seems to be similar to the results from older persons who have fragmented sleep; interestingly the
animal studies. association between elevated 24-h UFC and impaired sleep
Interestingly, recent studies have shown that GRs in and earlier awakening was seen in older women not receiving
humans are susceptible to epigenetic changes related to GR estrogen replacement therapy (196,197). Elevated interleukin
promoters in response to early-life experiences (35, 157, 181, 1-β also correlates with higher UFC levels (197). Levels of
222). Phenotypes with MR: GR imbalance due to genetic fac- UFC are found to be elevated in persons with dementia (145)
tors and early-life experiences might be vulnerable to late-life and memory decline (223).
stressors (183). Also Glucocorticoid Vulnerability Hypothesis When dynamic testing of the HPA axis with 1 mg
in contrast to the Glucocorticoid Cascade Hypothesis suggests overnight DEX suppression (1 mg orally administered at
that a history of chronic stress leaves an “imprint” on the HC, 2300) was performed, no significant difference in cortisol
making it vulnerable even when GCs are not elevated at the levels between young and healthy old subjects was seen;
time of metabolic challenge; hence, chronic stress and GCs however post DEX cortisol levels were higher in dementia
might play a critical role in priming HC to be more suscepti- patients compared to age-matched controls (143, 177). Some
ble to subsequent insults (41). More studies are needed in the studies have reported a high percentage of “non-responders”
future to validate this hypothesis. in healthy old subjects (about 30%) and in dementia patients
In young persons, cortisol has an early morning peak fol- (50%) (66, 67). Hence, it is thought that when aging is
lowed by a decline throughout the day reaching minimal associated with dementia, the adreno-cortical response to

1500 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

stressful events might be greater and more prolonged 67). Aging change Aging change
in males in females
It is well known that depression by itself can be associated
Cholesterol
with a pseudo-Cushingoid state. Intuitively, depressed elderly
(a common comorbidity) may demonstrate decreased ability
of DEX to suppress plasma cortisol and increased basal as Decreased Pregnenolone Decreased
well as stress-related cortisol secretion (32).
Pavlov et al. (191) examined the effects of an evening Decreased
Pregnenolone
sulfate Decreased
ovine CRH injection on stimulated cortisol, ACTH and DHEA
17 OH pregnenolone
levels in 49 healthy men, aged 21 to 86 years. Older men
had higher basal cortisol levels (P < 0.05) but there was
17,20-lyase(desmolase)
no difference in amplitude of either the ACTH or cortisol
response to ovine CRH. They, however, found that peak cor- Decreased Dehydroepiandrosterone Decreased
tisol responses occurred significantly earlier in older men.
DEX is less successful at blunting the CRH effect in older DHEA sulfate Decreased
Decreased
compared to younger persons (95). Conflicting results were
Androstenedione Decreased
seen when a psychosocial stress test elicited a higher ACTH Decreased
response in younger than older adults (127). More studies are Decreased Testosterone Decreased
needed to determine which of these studies is providing the Unchanged Estradiol Decreased
true pattern.
Unchanged 17 OH progesterone

11 Deoxycortisol

Basal Levels of Adrenal Steroid ? Unchanged Cortisol ? Unchanged

Hormones Unchanged Progesterone ? Unchanged

Four studies have examined the effects of aging on circulat- 11 Deoxycorticosterone


ing steroid hormones (13, 99, 131, 167). A composite of their
findings is shown in Figure 5. With aging there is a marked Corticosterone
decline in C-19 steroids [dehydroepiandrosterone (DHEA)
and DHEA-sulfate]. This leads to a decline in sex steroids. It Unchanged Aldosterone
is important to recognize these adrenal precursor steroids are
converted in active androgens and/or estrogens in peripheral Figure 5 The effect of aging on basal adrenal steroid hormones.
tissues (132). Thus, in males nearly half of the androgens
are derived in peripheral tissues from metabolizing enzymes
(133). In females approximately 100% of active estrogens
come from adrenal precursor steroids, and nearly three quar- increased ACTH secretion to overcome the decline in adrenal
ters prior to menopause (130). The decline in C-19 steroids corticosterone production. With aging there is an increase in
occurs predominantly between 20 to 30 years old and 50 to cholesterol ester stores in adrenal cells. This suggests that
60 years old. Changes beyond 60 years are less dramatic. there is impairment in cholesterol to undergo intramitochon-
Studies have shown a small decline in pregnenolone lev- drial transformation to pregnenolone.
els with age in men as well as women (13, 99, 131, 167). In Human studies with respect to adrenal responsiveness
addition, there is a marked decline in pregnenolone sulfate to ACTH have shown conflicting results. Maki (146) com-
after 60 years of age (99, 150, 167). The relatively intact lev- pared the effects of cosyntropin administration in young and
els of 17α OH progesterone, cortisol and progesterone with older males who had received DEX suppression. The 3 h
aging suggest that the 17, 20 lyase (desmolase) enzyme is age response of all adrenal hormones measured was lower in
sensitive. older males, despite an increase in the apparent half-life
of cosyntropin. At 24 h, there was an increase in 17-OH-
pregnenolone, providing further evidence for a decrease in
C17, 20 lyase function. While decreased C17, 20 lyase func-
Response of Aging Adrenals to ACTH tion was reported in two other studies (190, 250), these two
Old rats have a decline in ACTH-stimulated production of cor- studies showed unchanged or increased ACTH-stimulatability
ticosterone from adrenocortical cells (3). This is associated of cortisol, aldosterone, and other delta 4 steroids (androstene-
with an increase in the size of both the zona fasciculata and dione, 17-hydroxyprogesterone, and progesterone) with age
reticularis due to an increase in number and volume of cells than that in young subjects. The adrenal response of delta 5-
(130). Intracellularly, there is an increase in mitochondrial steroids (dehydroepiandrosterone, 17-hydroxypregnenolone,
volume and proliferation of the smooth endoplasmic reticu- and pregnenolone) to ACTH was found to be significantly
lum. These findings have been interpreted as an attempt by decreased with aging. We postulate that the above mentioned

Volume 4, October 2014 1501


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

Decreased The RAAS is one of the main systems involved in


glucocorticoid the regulation of blood pressure and sodium homeostasis.
receptor
Aging is associated with a decline in urinary excretion of
18-hydroxycortisol and aldosterone (235). In animal experi-
Mild ments and in humans, decreased plasma renin activity with a
cortisol
parallel lowering of aldosterone levels was observed with
aging. Young (20-30 year old) subjects were compared with
ACTH 11 Hydroxysteroid
elderly (62-70 year old) healthy subjects (254). The age-
Cortisone Cortisol related differences in plasma renin activity and aldosterone
Cholesterol Cortisone and were found to be more pronounced in stimulated conditions
esters poorly cortisol levels
converted to normal (sitting in an upright position, salt restriction, and plasma
pregnenolone
volume depletion) than under basal conditions (14, 102).
Age-related alterations of the kidney (glomerulosclerosis,
Increased numbers
and size of
Adipose decreased number of functional nephrons) might account for
tissue
glucocorticoid- increased the age-related differences in the active to inactive plasma
producing cells with aging renin ratio. A diminished synthesis of angiotensinogen by the
liver could contribute to the decreased activity of RAAS in
Triglycerides
absorbed aging. This is partially compensated by increased density of
postmeal angiotensin II receptors reported in elderly patients. The intra
renal system showed downregulation of renin mRNA and
Figure 6 Hypothesis: Cortisol increase in older persons is due to angiotensin-converting enzyme levels with aging, whereas
conversion of cortisone to cortisol in adipose tissue.
angiotensinogen levels remained stable. The decrease in
renin mRNA appeared to precede the fall in plasma renin
contradictory findings between the Maki and subsequent stud- concentration in the aging process. Additional studies in
ies could be due to DEX pretreatment in the Maki study. In 15-month-old rats confirmed that both basal and stimulated
another study, the adrenal androgen responsiveness to three renal renin release rates were impaired in older rats. Thus, both
doses of ACTH was examined (83). All three doses resulted decreased renin synthesis and impaired renin release underlie
in lower responses of DHEA to ACTH in older compared the fall in plasma renin with normal aging (119). Further-
to younger persons. The cortisol response to ACTH was more, aging is associated with a reduced adrenal responsive-
similar in older men compared to younger men at the two ness to angiotensin II, contributing to lower production of
higher doses. At the lowest dose there was a significantly aldosterone and alterations of sodium homeostasis. Adrenal
decreased cortisol response in the older group. To surmise, glomerulosa cells from old cows showed lower basal and
aging adrenals probably show decreased ACTH stimulation angiotensin stimulated aldosterone synthesis (195). Estradiol
of adrenal androgens but data with respect to ACTH effects and progesterone also stimulate the secretion of renin and
on other adrenal hormones is conflicting. reduced levels of these hormones at menopause could lead to
We postulate another reason for the small increases in reduced plasma renin activity.
circulating cortisol and the increased cortisol production rate Aging also has direct effects on adrenal cortical function.
seen in older persons. Just as androgenic steroids are produced There is 40% reduction of aldosterone in vivo in older rats
in peripheral tissues, so is cortisol from liver and adipose tis- given corticotropin-releasing hormone (27). A similar decline
sue by 11β hydroxysteroid dehydrogenase. It seems likely was seen in old rats after ACTH injection (3). In rats, isolated
that the increase in circulating cortisol in aging is due to an adrenal capsules containing predominantly the zona glomeru-
increase in adipose tissue associated with aging. This would losa, showed decreased basal secretion of aldosterone (200).
be compatible with the meal associated increase in cortisol These perfused capsules also demonstrated both a decreased
that has been seen in older persons, related to the increase in sensitivity and magnitude of response to ACTH and potas-
cortisol associated production in fat associated with increas- sium. This supports the fact that there is an age-related impair-
ing triglyceride deposition into fat (Fig. 6). ment in adrenal release of aldosterone. Older humans showed
a decrease in plasma response of 19 hydroxyandrost-4-ere-
3, 17 dione, and a lesser decrease in aldosterone to ACTH
Aldosterone stimulation (83, 107). Parker et al. (190) failed to show a
decrease in aldosterone production to ACTH in older humans.
Aldosterone is produced by the zona glomerulosa of the Morimoto et al. (165) reported a reduced aldosterone response
adrenal cortex. ACTH stimulates the secretion of aldos- to ACTH in sodium restricted older individuals. Overall, these
terone through the stimulation of deoxycorticosterone. Cir- studies support a small decrease in aldosterone secretion in
culating potassium levels also regulate aldosterone secretion. response to ACTH with aging.
The major regulator of aldosterone is the renin-angiotensin- Hyperaldosteronism is the most common cause of sec-
aldosterone system (RAAS). ondary hypertension. It occurs in between 5% and 15% of

1502 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

middle aged or older males. Hyperaldosteronism is the most alternatively, changes in receptor sensitivity might be the early
common cause of resistant hypertension (194). These per- event that leads to a compensatory increase in sympathetic
sons tend to be older, have greater vascular stiffness and pro- nerve activity (110).
nounced left ventricular hypertrophy. Many of these persons
have bilateral adrenal hyperplasia, most probably secondary
to the age-associated increase in ACTH secretion. Recently, Frailty and Corticosteroids
a subset of these individuals has been identified with gain of
function mutations in KLNJ5 (6). These patients have small Frailty is defined as an older person who is functioning nor-
aldosterone producing adenomas, often less than 1 cm, that mally, but when exposed to stressful conditions (either phys-
are missed with computed tomography. Again, expression of ical or psychological) has a decreased ability to maintain
these mutations may be secondary to the age-related increase his/her activities of daily living (166, 169). Frailty is a pre-
in ACTH secretion, particularly as these cells anatomically disability state and as such needs to be distinguished from
resemble cortisol secreting cells of the zona fasciculate. disability.
In 2001, Fried and her colleagues (74) created a physi-
cal frailty phenotype. This consisted of weight loss (10 lbs.
in 1 year), exhaustion (by self-report), grip strength (in the
Adrenomedullary Hormones lowest 20%), walking speed (in the lowest 20%), and low
physical activity (kcals/week in the lowest 20%). This phys-
and Aging ical phenotype has been shown to be highly predictive of the
Central administration of CRH causes increases in development of disability and mortality (8,23). Epidemiolog-
epinephrine and norepinephrine, making it the coordinator ical studies have suggested a number of endocrine changes
of the peripheral responses to stress (26). In rats, there is a with aging including vitamin D deficiency and low testos-
decrease in CRH mRNA levels in the paraventricular nucleus terone (in males) are related to frailty (31, 60, 259).
with aging and this is related to a decline in catecholamine The International Association of Nutrition and Aging has
response to stress (39). developed an extremely simple screen for frailty—the FRAIL
With aging, sympathetic activation was evident in the (1, 2). This has been shown to have similar predictive failure
heart, gut and the liver at rest as well as in response to imme- to other frailty screening tests (168, 260). The components of
diate as well as prolonged stressors. Sympathetic nerve activ- the FRAIL are given in Table 2.
ity at rest, also known as sympathetic tone, is found to be Frail persons are particularly vulnerable to stress and
increased in many parts of the body during aging (63, 248). so it has been postulated that increased activity of the
The mechanism is thought to be increased turnover of sym- hypothalamic-pituitary-adrenal axis may accelerate their
patho excitatory noradrenaline (or norepinephrine) accompa- decline. Previously, it has been shown that psychosocial stres-
nied by activation of sympathetic outflows to the heart and sors, for example, the period after earthquakes or the decision
hepatomesenteric circulation but not to the kidneys (64, 86). of Britain to return Hong Kong to China, can increase mor-
Conversely, adrenal medullary release of epinephrine is sub- tality in old-old persons (36,70). In an attempt to confirm this
normal in the elderly, at rest and during stress (62). It was hypothesis, a number of studies on the hypothalamic-pituitary
found that adrenaline (or epinephrine) secretion from the adrenal axis have been undertaken in frail older persons.
adrenal medulla was 40% lower in older compared with young Utilizing community dwelling subjects in the Women’s
healthy men (248). However, this difference was not reflected Health and Aging Study, it was found that both evening corti-
in corresponding plasma adrenaline (or epinephrine) concen- sol and 24 h salivary cortisol were positively correlated with
tration, which is not significantly different based on age, frailty burden (247). Frailty burden was defined by using the
as plasma clearance was 20% lower in older men. Finally, Fried physical frailty phenotype. Frailty burden was also asso-
adrenaline might be released from the heart at rest in older ciated with a smaller circadian variation in cortisol. Holanda
individuals. The postulated mechanisms for decreased adrenal et al. (109) also reported increased salivary cortisol in frail
adrenaline production are reductions in preganglionic nerve persons. Another study in nursing home residents found that
activity to the adrenal medulla and reduction in adrenaline frail individuals were suppressed to a lesser degree by DEX
synthesis and storage in the adrenal medulla. Thus, the age- than were more successful agers (33). In the Hertfordshire
related changes in sympathetic nerve activity appear to vary
depending on the particular organ system (212, 221). Data
Table 2 Components of a Simple Screening Test for Frailty—the
indicate the changes at the effector level; for example, vaso- FRAIL
constriction and tachycardia in response to standing are the
result of changes in catecholamine receptor function on end-
Fatigue
organ tissues and impairments in end-organ function occur Resistance (climb 1 flight of stairs)
despite well-maintained or even increased sympathetic nerve Aerobic (walk one block)
activity. This decline of catecholamine receptor sensitivity Illness (more than 5)
Loss of weight (>5% in 6 months)
during aging might be a result of receptor desensitization;

Volume 4, October 2014 1503


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

Aging Study, a high cortisol: DHEA sulfate ratio was associ- mechanisms behind cortisol elevation could be dysfunction
ated with increased frailty (12). of MRs in HC with aging. As mentioned above, these high-
In addition to these findings, Rao et al. (201) found that affinity receptors bind cortisol at low levels in the HC and
only 25% of frail patients responded to 1 mcg ACTH stim- hence control feedback inhibition on HPA-axis at the nadir
ulation compared to 80% of nonfrail controls. This relative of the circadian rhythm. Salivary cortisol studies seemed to
hypoadrenalism in frail compared to other older persons needs support these results. The reason for the failure to find an
to be studied further but there is no current evidence to estab- increase in 24 h urine cortisol is uncertain, but could be due
lish a causal relationship between hypoadrenalism and frailty. to renal dysfunction for aging. While the glucocorticoid cas-
The effect on cortisol changes with aging has also been case hypothesis proposed chronically elevated GC levels in
studied on physical performance tests. In the Whitehall II aged mice leading to neuronal cell loss in the HC resulting in
and Caterphilly Prospective Cohort slower walking speed memory loss, these findings could not be replicated in other
was associated with a reduction in diurnal cortisol variabil- animal models and humans making us question the validity
ity (79, 129). In the Longitudinal Aging Study Amsterdam of this hypothesis. High GCs in aging humans could prime
(LASA), women with higher cortisol levels showed worse per- the aging brain to future metabolic stressors as suggested in
formance on a standing balance test; while men had a slower the glucocorticoid vulnerability hypothesis, but more studies
walking speed and were slower to do chair rises (192). Lower are needed in this area before making any conclusions.
diurnal salivary cortisol levels were associated with poorer While ACTH levels in aging are also increased, there
performance on the Berg Balance Scale and a decrease in could be decreased adrenal responsiveness to ACTH; studies
handgrip performance (100). In this study, older participants show conflicting data with respect to cortisol but there seems
had a significantly higher cortisol area under the curve (AUC), to be decreased adrenal androgen production in response to
lower overall DHEA levels, lower DHEA AUC, a decreased ACTH. Decreased C 17, 20 lyase levels have been reported.
diurnal slope of decline and increased cortisol: DHEA ratio. It is possible that much of the increase in cortisol with aging
Sarcopenia is defined as the loss of muscle mass asso- is due to conversion of cortisone to cortisol in adipose tissue,
ciated with limited mobility (68, 208). Miller et al. (162) which is increased with aging; however, this is our hypothesis
examined younger patients with computed tomography deter- and there is no literature to support this.
mined loss of psoas muscle area and fat infiltration. They The steroid precursor, pregnenolone, is decreased with
compared them to age matched controls without muscle loss. aging due to problems with lipid uptake into mitochon-
They found that both muscle loss and fat infiltration was asso- dria. The C19 steroids (DHEA and DHEA-S) are markedly
ciated with increased 24 urine cortisol secretion. Waters et al. decreased with aging; however the clinical significance of
(253) reported elevated cortisol and leptin levels in sarcopenic this is questionable. Aldosterone shows a small decrease and
older subjects relative to their low body fat mass. Overnight adrenal medullary epinephrine secretion declines with aging.
higher UFC levels were an independent predictor of future Overall, conflicting studies and the problem of reconciling
fracture at any site (88). in vivo and in vitro studies and the decline in glucocorticoid
DHEA and its sulfate have been touted as antiaging receptor activity with aging makes it difficult to draw firm
hormones. Again, literature shows conflicting data. DHEA- conclusions concerning the physiological outcomes of alter-
S has been shown epidemiologically to predict frailty ations in the HPA axis with aging. The role, if any, of the HPA
(68, 136, 208, 251). Valenti et al. (242) found that in males axis in aging, frailty, sarcopenia and Alzheimer’s is uncertain
aged 60 to 79 years of age, DHEA-S was correlated with at this time.
muscle strength and calf muscle area. In young menopausal
women DHEA-S was related to loss of appendicular muscle
mass but not muscle strength (209). Lower DHEA-S levels
were associated with a higher degree of physical disability in References
women (92). However, in middle aged males DHEA-S failed 1. Abellan van Kan G, Rolland Y, Bergman H, Morley JE, Kritchevsky
to correlate with either muscle mass or grip and lower limb SB, Vellas B. The I.A.N.A. Task Force on frailty assessment of older
people in clinical practice. J Nutr Health Aging 12: 29-37, 2008.
muscle strength (91). Replacement studies of DHEA on mus- 2. Abellan van Kan G, Rolland YM, Morley JE, Vellas B. Frailty: Toward
cle mass and strength have had minimal effects, questioning a clinical definition. J Am Med Dir Assoc 9: 71-72, 2008.
3. Ait-Chaoui A, Rakotondrazafy J, Brudieux R. Age-related changes in
DHEA as a likely antiaging hormone (7, 126). plasma corticosterone and aldosterone responses to exogenous ACTH
in the rat. Horm Res 43: 181-187, 1995.
4. Antoni FA. Hypothalamic control of adrenocorticotropin secretion:
Advances since the discovery of 41-residue corticotropin-releasing fac-
Conclusion 5.
tor. Endocr Rev 7: 351-378, 1986.
Arvat E, Baccagno B, Giordano R, Pellegrino M, Broglio F, Gianotti L,
Maccario M, Camanni F, Ghigo E. Mineralocorticoid receptor block-
There is reasonable evidence to conclude that with aging there ade by canrenoate increases both spontaneous and stimulated adrenal
is an increase in cortisol at the circadian trough and possi- function in humans. J Clin Endocrinol Metab 86: 3176-3181, 2001.
6. Azizan EA, Poulsen H, Tuluc P, Zhou J, Clausen MV, Lieb A, Maniero
bly in total throughout the day. There is a clear blunting in C, Garg S, Bochukova EG, Zhao W, Shaikh LH, Brighton CA, Teo
the diurnal rhythm of cortisol production as well as inade- AE, Davenport AP, Dekkers T, Tops B, Küsters B, Ceral J, Yeo GS,
Neogi SG, McFarlane I, Rosenfeld N, Marass F, Hadfield J, Margas W,
quate cortisol suppression with DEX suppression. One of the Chaggar K, Solar M, Deinum J, Dolphin AC, Farooqi IS, Striessnig J,

1504 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

Nissen P, Brown MJ. Somatic mutations in ATP1A1 and CACNA1D 30. Cannon W. Bodily Changes in Pain, Hunger, Fear, and Rage. By Walter
underlie a common subtype of adrenal hypertension. Nat Genet 345: B. Cannon. 404 pages. New York: D. Appleton & Co., 1929, pp. 219-
1055-1060, 2013. 230. Reviewed by Karl M. Bowman Am J Psychiatry 1930; 86: 770-771.
7. Baker WL, Karan S, Kenny AM. Effect of dehydroepiandrosterone on 31. Carcaillon L, Blanco C, Alonso-Bouzon C, Alfaro-Acha A, Garcia-
muscle strength and physical function in older adults: A systematic Garcia FJ, Rodriguez-Manas L. Sex differences in the association
review. J Am Geriatr Soc 59: 997-1002, 2011. between serum levels of testosterone and frailty in an elderly popu-
8. Bandeen-Roche K, Xue QL, Ferrucci L, Walston J, Guralnik JM, lation: The Toledo Study for Healthy Aging. PLoS One 7(3):e32401,
Chaves P, Zeger SL, Fried LP. Phenotype of frailty: Characterization 2012.
in the women’s health and aging studies. J Gerontol A Biol Sci Med Sci 32. Carroll BJ, Feinberg M, Greden JF, Tarika J, Albala AA, Haskett RF,
61: 262-266, 2006. James NM, Kronfol Z, Lohr N, Steiner M, de Vigne JP, Young E. A
9. Barton RN, Horan MA, Clague JE, Rose JG. The effect of aging on specific laboratory test for the diagnosis of melancholia: Standardiza-
the metabolic clearance rate and distribution of cortisol in man. Arch tion, validation and clinical utility. Arch Gen Psychiatry, 38: 15-22,
Gerontol Geriatr 29: 95-105, 1999. 1981.
10. Barton RN, Horan MA, Weijers JW, Sakkee AN, Roberts NA, van 33. Carvalhaes-Neto N, Huayllas MK, Ramos LR, Cendoroglo MS, Kater
Bezooijen CF. Cortisol production rate and the urinary excretion of CE. Cortisol, DHEAS and aging: Resistance to cortisol suppression in
17-hydroxycorticosteroids, free cortisol, and 6 beta-hydroxycortisol in frail institutionalized elderly. J Endocrinol Invest 26: 17-22, 2003.
healthy elderly men and women. J Gerontol 48:M213-M218, 1993. 34. Carvalhaes-Neto N, Ramos LR, Vieira JG, Kater CE. Urinary free
11. Bateman A, Singh A, Kral T, Solomon S. The immune-hypothalamic- cortisol is similar in older and younger women. Exp Aging Res 28:
pituitary-adrenal axis. Endocr Rev 10: 92-112, 1989. 163-168, 2002.
12. Baylis D, Bartlett DB, Syddal HE, Ntani G, Gale CR, Cooper C, Lord 35. Champagne FA, Francis DD, Mar A, Meaney MJ. Variations in maternal
JM, Sayer AA. Immune-endocrine biomarkers as predictors of frailty care in the rat as a mediating influence for the effects of environment
and mortality: A 10-year longitudinal study in community-dwelling on development. Physiol Behav 79: 359-371, 2003.
older people. Age (Dordr) 35: 963-971, 2013. 36. Chau PH, Yen E, Morley JE, Woo J. The effects of environmental
13. Belanger A, Candas B, Dupont A, Cusan L, Diamond P, Gomez JL, stressors on the mortality of the oldest old male population in Hong
Labrie F. Changes in serum concentrations of conjugated and unconju- Kong, 1977-2006. Aging Male 11: 179-188, 2008.
gated steroids in 40- to 80-year-old men. J Clin Endocrinol Metab 79: 37. Chen R, Lewis KA, Perrin MH, Vale WW. Expression cloning of
1086-1090, 1994. a human corticotropin-releasing-factor receptor. Proc Natl Acad Sci
14. Belmin J, Levy BI, Michel JB. Changes in the renin-angiotensin- U S A 90: 8967-8971, 1993.
aldosterone axis in later life. Drugs Aging 5: 391-400, 1994. 38. Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-
15. Berardelli R, Karamouzis I, D’Angelo V, Berardelli R, Karamouzis J, mediated inflammation. N Engl J Med 332: 1351-1362, 1995.
D’Angelo V, Zichi C, Fussotto B, Giordano R, Ghigo E, Arvat E. Role 39. Cizza G, Gold PW, Chrousos GP. Aging is associated in the 344/N
of mineralocorticoid receptors on the hypothalamus-pituitary-adrenal Fischer rat with decreased stress responsivity of central and periph-
axis in humans. Endocrine 43: 51-58, 2013. eral catecholaminergic systems and impairment of the hypothalamic-
16. Berardelli R, Karamouzis I, Marinazzo E, Prats E, Picu A, Giordano pituitary-adrenal axis. Ann NY Acad Sci 771: 491-511, 1995.
R, Ghigo E, Arvat E. Effect of acute and prolonged mineralocorti- 40. Collaziol D, Luz C, Dornalles F, da Cruz IM, Bauer ME. Psychoneu-
coid receptor blockade on spontaneous and stimulated hypothalamic- roendocrine correlates of lymphocyte subsets during healthy ageing.
pituitary-adrenal axis in humans. Eur J Endocrinol 162: 1067-1074, Mech Ageing Dev 125: 219-27, 2004.
2010. 41. Conrad CD. Chronic stress-induced hippocampal vulnerability: The
17. Berger S, Wolfer Dp, Selbach O, Alter H, Erdmann G, Reichardt HM, glucocorticoid vulnerability hypothesis. Rev Neurosci 19: 395-411,
Chepkova AN, Welzl H, Haas HL, Lipp HP, Schutz G. Loss of the 2008.
limbic mineralocorticoid receptor impairs behavioral plasticity. Proc 42. Dallman MF, Levin N, Cascio CS, Akana SF, Jacobson L, Kuhn
Natl Acad Sci USA 103: 95-200, 2006. RW. Pharmacological evidence that the inhibition of diurnal adreno-
18. Bizon JL, Helm KA, Han JS, Chun HJ, Pucilowska J, Lund PK, Gal- corticotropin secretion by corticosteroids is mediated via type I
lagher M. Hypothalamic-pituitary-adrenal axis function and corticos- corticosterone-preferring receptors. Endocrinology 124: 2844-2850,
terone receptor expression in behaviourally characterized young and 1989.
aged Long-Evans rats. Eur J Neurosci 14: 1739-1751, 2001. 43. Dalm S, Enthoven L, Meijer OC, Van der Mark MH, Karssen AM, de
19. Born J, de Kloet ER, Wenz H, Kern W, Fehm HL. Gluco- and antimin- Kloet ER, Oitzl MS. Age-related changes in hypothalamic-pituitary-
eralocorticoid effects on human sleep: A role of central corticosteroid adrenal axis activity of male C57BL/6J mice. Neuroendocrinology 81:
receptors. Am J Physiol 260: 183-188, 1991. 372-380, 2005.
20. Born J, Ditchuneit I, Schreiber M, Dodt C, Fehm HL. Effects of age 44. Davis LG, Arentzen R, Reid JM, Manning RW, Wolfson B, Lawrence
and gender on pituitary-adrenocortical responsiveness in humans. Eur KL, Baldino F Jr. Glucocorticoid sensitivity of vasopressin mRNA
J Endocrinol 132: 705-711, 1995. levels in the paraventricular nucleus of the rat. Proc Natl Acad Sci
21. Born J, Steinbach D, Dodt C, Fehm HL. Blocking of central nervous U S A 83: 1145-1149, 1986.
mineralocorticoid receptors counteracts inhibition of pituitary-adrenal 45. de Kloet ER, Joels M, Holsboer F. Stress and the brain: From adaptation
activity in human sleep. J Clin Endocrinol Metab 82: 1106-1110, to disease. Nature Rev Neurosci 6: 463-475, 2005.
1997. 46. de Kloet ER, Oitzl ER, Vermetten E. Stress hormones and PTSD: Basic
22. Boscaro M, Paoletta A, Scarpa E, Barzon L, Fusaro P, Fallo F, Soniero studies and clinical perspectives. Prog Brain Res 167: 1-320, 2008.
N. Age-related changes in glucocorticoid fast feedback inhibition of 47. de Kloet ER, Vreugdehil E, Oitzl MS, Joels M. Brain corticos-
adrenocorticotropin in man. J Clin Endocrinol Metab 83: 1380-1383, teroid receptor balance in health and disease. Endocr Rev 9: 269-301,
1998. 1998.
23. Boyd CM, Xue QL, Simpson CF, Guralnik JM, Fried LP. Frailty, 48. Dek Michalska AG, Spyrka J, Rachwalska P, Tadeusz J, Bugajski J.
hospitalization, and progression of disability in a cohort of disabled Influence of chronic stress on brain corticosteroid receptors and HPA
older women. Am J Med 118: 1225-1231, 2005. axis activity. Pharmacol Rep 65: 1163-1175, 2013.
24. Boyle MP, Brewer JA, Funatsu M, Qozniak DF, Tsien JZ, Isumi Y, 49. de Pablos RM, Villáran RF, Argüelles S, Herrera AJ, Venero JL, Ayala
Muglia LJ. Acquired deficit of forebrain glucocorticoid receptor pro- A, Cano J, Machado A. Stress increases vulnerability to inflammation
duces depression-like changes in adrenal axis regulation nd behavior. in the rat prefrontal cortex. J Neurosci 26: 5709-5719, 2006.
Proc Natl Acad Sci USA 102: 473-478, 2005. 50. Deuschle M, Gotthardt U, Schweiger U, Weber B, Korner A, Schmider
25. Brinks V, van der Mark MH, de Koet ER, Oitzl MS. Differential MR/GR J, Standhardt H, Lammers Ch, Heuser I. With aging in humans the
activation in mice results in emotional states beneficial or impairing for activity of the hypothalamus-pituitary-adrenal system increases and its
cognition. Neural Plast, 2007: 90163, 2007. diurnal amplitude flattens. Life Sci 61: 2239-2246, 1997.
26. Brown MR, Fisher LA, Spiess J, Rivier C, Rivier J, Vale W. 51. Deuschle M, Weber B, Colla M, Muller M, Kniest A, Heuser I. Miner-
Corticotropin-releasing factor: Actions on the sympathetic nervous sys- alocorticoid receptor also modulates basal activity of hypothalamus-
tem and metabolism. Endocrinology 111: 928-931, 1982. pituitary-adrenocortical system in humans. Neuroendocrinology 68:
27. Brudieux R, Ait Chaoui A, Rakotondrazafy J. Age-related decreases in 355-360, 1998.
plasma adrenocorticotropic hormone, corticosterone, and aldosterone 52. Dinkel K, MacPherson A, Sapolsky RM. Novel glycocorticoid effects
responses to exogenous corticotropin-releasing hormone in the rat. on acute inflammation in the CNS. J Neurochem 84: 705-716, 2003.
Gerontology 41: 308-314, 1995. 53. Djardjevic-Markovic R, Radic O, Jelic V, Radojcic M, Rapie-Otnir V,
28. Buckley TM, Mullen BC, Schatzberg AF. The acute effects of a min- Ruzdijcs S, Krstic-Demonaios M, Kanazir S, Kanazir D. Glucocorticoid
eralocorticoid receptor (MR) agonist on nocturnal hypo- thalamic- receptors in aging rats. Exp Gerontol 34: 971-982, 1999.
adrenal-pituitary (HPA) axis activity in healthy controls. Psychoneu- 54. Dmitrieva NO, Almeida DM, Dmitrieva J, Loken E, Pieper CF. A day-
roendocrinology 32: 859-864, 2007. centered approach to modeling cortisol: Diurnal cortisol profiles and
29. Bush IE, Sandberg AA. Adrenocortical hormones in human plasma. their associations among U.S. adults. Psychoneuroendocrinology 38:
J Biol Chem 205: 783-793, 1953. 2354-2365, 2013.

Volume 4, October 2014 1505


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

55. Dodt C, Dittman J, Hruby J, Spath-Schwalbe E, Born J, Schuttler 77. Gaillard RC, Al Damluji S. Stress and pituitary-adrenal axis. Bailliere
R, Fehm HL. Different regulation of adrenocorticotropin and cortisol Clin Endocrinol Metab 1: 319-354, 1987.
secretion in young, mentally healthy elderly, and patients with senile 78. Garrido P. Aging and stress: Past hypothesis, present approaches and
dementia of Alzheimer’s type. J Clin Endocrinol Metab 72: 272-276, perspectives. Aging Dis 2: 80-99, 2011.
1991. 79. Gardner MP, Lightman SL, Gallacher J, Hardy R, Kuh D, Ebrahim
56. Dodt C, Kern W, Fehm HL, Born J. Antimineralocorticoid can- S, Boyer A, Ben-Shlomo Y, Halcyon Study Team. Diurnal cortisol
renoate enhances secretory activity of the hypothalamus-pituitary- patterns are associated with physical performance in the Caerphilly
adrenocortical (HPA) axis in humans. Neuroendocrinology 58: 570- Prospective Study. Int J Epidemiol 40: 1693-1702, 2011.
574, 1993. 80. Gardner MP, Lightman S, Sayer AA, Coopoer C, Cooper R, Deeg D,
57. Dodt C, Theine KJ, Uthgennant D, Born J, Fehm HL. Basal secretory Ebrahim S, Gallacher J, Kivimaki M, Kumari M, Kuh D, Martin Rm,
activity of the hypothalamo-pituitary-adrenocortical axis is enhanced Peeters G, en-Schlomo Y; Halcyon Study Team. Dysregulation of the
in healthy elderly. An assessment during undisturbed night-time sleep. hypothalamic pituitary adrenal (HPA) axis and physical performance
Eur J Endocrinol 13: 443-450, 1994. at older ages: An individual participant meta-analysis. Psychoneuroen-
58. Donald RA. ACTH and related peptides. Clin Endocrinol (Oxf.) 12: docrinology 38: 40-49, 2013.
491-524, 1980. 81. Ginsberg AB, Campeau S, Day HE, Spencer RL. Acute glucocorticoid
59. Dong Y, Poellinger L, Okret S, Hoog JO, von Bahr-Lindstrom H, pretreatment suppresses stress-induced hypothalamic-pituitary-adrenal
Jornvall H, Gustafsson JA. Regulation of gene expression of class I axis hormone secretion and expression of corticotropin-releasing hor-
alcohol dehydrogenase by glucocorticoids. Proc Natl Acad Sci USA mone hnRNA but does not affect c-fox mRNA or fox protein expression
85: 767-771, 1988. in the paraventricular nucleus of the hypothalamus. J Neuroendocrinol
60. Eichholzer M, Barbir A, Basaria S, Dobs AS, Feinleib M, Guallar 15: 1075-1083, 2003.
E, Menke A, Nelson WG, Rifai N, Platz EA, Rohrmann S. Serum 82. Giordono R, Bo M, Pallegrino M, Vessari M, Baldi M, Pieu A, Balbo
sex steroid hormones and frailty in older American men of the Third M, Bonelli L, Migliaretti G, Ghigo E, Arvat E. Hypothalamus-pituitary-
National Health and Nutrition Examination Survey (NHANES III). adrenal hyperactivity in human aging is partially refractory to stimu-
Aging Male 15: 208-215, 2012. lation by mineralcorticoid receptor blockade. J clin Endocrniol Metab
61. Erickson K, Drevets W, Schulkin J. Glucocorticoid regulation of diverse 90: 5656-5662, 2005.
cognitive functions in normal and pathological emotional states. Neu- 83. Giordano R, Di Vito L, Lanfranco F, Broglio F, Beuso A, Gianotti L,
rosci Biobehav Rev 3: 233-246, 2003. Grottoli S, Ghigo E, Arvat E. Elderly subjects show severe impairment
62. Esler M, Hastings J, Lambert G, Kaye D, Jennings G, Seals DR. of dehydroepiandrosterone sulphate and reduced sensitivity of cortisol
The influence of aging on the human sympathetic nervous system and aldosterone response to the stimulatory effect of ACTH (1-24).
and brain norepinephrine turnover. Am J Physiology 282:R909-R916, Clin Endocrinol (Oxf) 55: 259-265, 2001.
2002. 84. Goncharova ND. Stress responsiveness of the hypothalamic-pituitary-
63. Esler M, Lambert G, Kaye D, Rumantir M, Hastings J, Seals DR. adrenal axis: Age-related features of the vasopressinergic regulation.
Influence of ageing on the sympathetic nervous system and adrenal Front Endocrinol (Lausanne) 4: 26ECollection, 2013.
medulla at rest and during stress. Biogerontology 3: 45-49, 2002. 85. Grad B, Kral UA, Payne RC, Berenson J. Plasma and urinary corticoids
64. Esler MD, Turner AG, Kaye DM, Thompson JM, Kingwell BA, Morris in young and older persons. J Gerontol 22: 66-71, 1967.
M, Lambert GW, Jennings GL, Cox HS, Seals DR. Aging effects on 86. Grassi M, Esler M. How to assess sympathetic activity in humans.
human sympathetic neuronal function. Am J Physiol 268(1 Pt 2): R278- J Hypertens 17: 719-734, 1999.
R285, 1995. 87. Greendale GA, Kritz-Silverstein D, Seeman T, Barrett-Connor E.
65. Ferrari E, Casarotti D, Muzzoni B, Albertelli N, Cravello L, Fioravanti Higher basal cortisol predicts verbal memory loss in postmenopausal
M, Solerte SB, Magri F. Age-related changes of the adrenal secretory women: Rancho Bernardo Study. J Am Geriatr Soc 48: 1645-1658,
pattern: Possible role in pathological brain aging. Brain Res Rev 37: 2000.
294-300, 2001. 88. Greendale GA, Unger JB, Rowe JW, Seeman TE. The relation between
66. Ferrari E, Cravallo L, Muzzoni B, Casarotti D, Paltro M, Solerte SB, cortisol excretion and fractures in healthy older people: Results from
Fioravanti M, Cuzzoni G, Pontigsia B, Magri F. Age-related changes of the MacArthur studies-Mac. J Am Geriatr Soc 47: 799-803, 1999.
the hypothalamic-pituitary-adrenal axis: Pathophysiological correlates. 89. Grottoli S, Giordano R, Maccagno B, Pellegrino M, Ghigo E, Arvat E.
Eur J Endocrinol, 144: 319-29, 2001. The stimulatory effect of canrenoate, a mineralocorticoid antagonist,
67. Ferrari M, Mantero F. Male aging and hormones: The adrenal cortex. on the activity of the hypothalamus-pituitary-adrenal axis is abolished
J Endocrinol Invest 28: 92-95, 2005. by alprazolam, a benzodiazepine, in humans. J Clin Endocrinol Metab
68. Fielding RA, Vellas B, Evans WJ, Bhasin S, Morley JE, Newman 87: 4616-4620, 2002.
AB, Abellan van Kan G, Andrieu S, Bauer J, Breuille D, Cederholm T, 90. Gust DA, Wilson ME, Stocker T, Conrad S, Plotsky PM, Gordon
Chandler J, De Maynard C, Donini L, Harris T, Kannt A, Keime Guibert TP. Activity of the hypothalamic-pituitary-adrenal axis is altered by
F, Onder G, Papanicolaou D, Rolland Y, Rooks D, Sieber C, Souhami aging and exposure to social stress in female rhesus monkeys. J Clin
E, Verlaan S, Zamboni M. Sarcopenia: An undiagnosed condition in Endocrinol Metab 85: 2556-2563, 2000.
older adults. Current consensus definition: Prevalence, etiology, and 91. Haren MT, Banks WA, Perry III HM, Patrick P, Malmstrom TK,
consequences. International Working Group on Sarcopenia. J Am Med Miller DK, Morley JE. Predictors of serum testosterone and DHEAS
Dir Assoc 12: 249-256, 2011. in African-American men. Int J Androl 31: 50-59, 2008.
69. Fish HR, Chernow B, O’Brian JT. Endocrine and neurophysiologic 92. Haren MT, Malmstrom TK, Banks WA, Patrick P, Miller DK, Morley
responses of the pituitary to insulin-induced hypoglycemia: A review. JE. Lower serum DHEAS levels are associated with a higher degree of
Metabolism 35: 763-780, 1986. physical disability and depressive symptoms in middle-aged to older
70. Flaherty JH, Dong B, Wu H, Zhang Y, Guralnik JM, Malmstrom TK, African American women. Maturitas 57: 347-360, 2007.
Morley JE. Observational study of 1-year mortality rates before and 93. Harris AP, Holmes MC, de Kloet ER, Chapman KE, Seckl JR.
after a major earthquake among Chinese nonagenarians. J Gerontol A Mineralo-corticoid and glucocorticoid receptor balance in control
Biol Sci Med Sci 66: 355-361, 2011. of HPA axis and behavior. Psychoneuroendocrinology 28: 648-658,
71. Franceschi M, Airaghi L, Gramigna C, Truci G, Manfedi MG, Canal N, 2013.
Catnaia A. ACTH and cortisol secretion in patients with Alzheimer’s 94. Harris BN, Saltzman W. Effects of aging on hypothalamic-pituitary-
disease. J Neurol Neurosurg Psychiatry 54: 836-837, 1991. adrenal (HPA) axis activity and reactivity in virgin male and female
72. Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. California mice (Peromyscus californicus). Gen Comp Endocrinol 186:
Microglia serve as a neuroimmune substrate for stress-induced poten- 41-49, 2013.
tiation of CNS pro-inflammatory cytokines responses. Brain Behav 95. Hatzinger M, Brand S, Herzig N, Holsboer-Trachsler E. In healthy
Immun 21: 47-59, 2007. young and elderly adults, hypothalamic-pituitary-adrenocortical axis
73. Frank MG, Miguel ZD, Watkins LR, Maier SF. Prior exposure to glu- reactivity (HPA AR) varies with increasing pharmacological challenge
cocorticoids sensitizes the neuroinflammatory and peripheral inflam- and with age, but not with gender. J Psychiatr Res 45: 1373-1380,
matory responses to E. Coli lipopolysaccharide. Brain Behav Immun 2011.
24: 19-30, 2010. 96. Hatzinger M, Reul JM, Landgraf R, Holsboer F, Neumann I.
74. Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Combined dexamethasone/CRH test in rats: Hypothalamo-pituitary-
Seeman T, Tracy R, Kop WJ, Burke G, McBurnie MA; Cardiovascular adrenocortical system alterations in aging. Neuroendocrinology 64:
Health Study Collaborative Research Group. Frailty in older adults: 349-356, 1996.
Evidence for a phenotype. J Gerontol A Biol Sci Med Sci 56:M146- 97. Hauger RL, Aguilera G. regulation of pituitary corticotropin releas-
M156, 2001. ing hormone (CRH) receptors by CRH: Interaction with vasopressin.
75. Fuchs E, Flugge G. Chronic social stress effects on limbic brain struc- Endocrinology 133: 1708-1714, 1993.
tures. Physiol Behav 79: 417-427, 2003. 98. Hauger RLM, Thrivikraman KV, Plotsky PM. Age-related alterations
76. Funder JW. Glucocorticoid and mineral corticoid receptors: Biology of hypothalamic-pituitary-adrenal axis function in male Fischer 344
and clinical relevance. Annu Rev Med 48: 231-240, 1997. rats. Endocrinology 134: 1528-1536, 1994.

1506 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

99. Havlikova H, Hill M, Hampl R, Starka L. Sex- and age-related changes 2001. http://www.nia.gov/sites/default/files/AgingWorld2001_0.pdf.
in epitestosterone in relation to pregnenolone sulfate and testosterone Accessed October 29, 2013.
in normal subjects. J Clin Endocrinol Metab 87: 2225-2231, 2002. 126. Kritz-Silverstein D, von Muhlen D, Laughlin GA, Bettencourt R.
100. Heaney JL, Phillips AC, Carroll D. Ageing, physical function, and the Effects of dehydroepiandrosterone supplementation on cognitive
diurnal rhythms of cortisol and dehydroepiandrosterone. Psychoneu- function and quality of life: The DHEA and Well-Ness (DAWN) Trial.
roendocrinology 37: 341-349, 2012. J Am Geriatr Soc 56: 1292-1298, 2008.
101. Herman JP, Figueiredo H, Mueller NK, Ulrich-Lai Y, Ostran- 127. Kudielka BM, Buske-Kirschbaum A, Hellhammer DH, Kirschbaum
der MM, Choi DC, Cullinau WE. Central mechanisms of stress C. HPA axis responses to laboratory psychosocial stress in healthy
integration: Hierarchical circuitry controlling hypothalamo-pituitary- elderly adults, younger adults, and children: Impact of age and gender.
adrenocortical responsiveness. Front Neuroendocrinol, 24: 151-180, Psychoneuroendocrinology, 29: 83-98, 2004.
2003. 128. Kumari M, Badrick E, Chandola T, Adler NE, Epel E, Seeman T,
102. Hegstad R, Brown RD, Jiang NS, Kao P, Weinshilboum RM, Strong Kirschbaum D, Marmot MG. Measures of social position and cortisol
C, Wisgerhof M. Aging and aldosterone. Am J Med 74: 442-448, secretion in an aging population: Findings from the Whitehall II study.
1983. Psychosom Med 72: 27-34, 2010.
103. Herman JP, Larson BR, Speert DB, Seasholtz AF. Hypothalamo- 129. Kumari M, Badrick E, Sacker A, Kirschbaum C, Marmot M, Chandola
pituitary-adrenocortical dysregulation in aging F344/Brown-Norway T. Identifying patterns in cortisol secretion in an older population.
F1 hybrid rats. Neurobiol Aging 22: 323-332, 2001. Findings from the Whitehall II study. Psychoneuroendocrinology, 35:
104. Herman JP, Schafer MK, Young EA, Thompson R, Douglass J, Akil 1091-1099, 2010.
H, Watson SJ. Evidence for hippocampal regulation of neuroendocrine 130. Labrie F. Intracrinology. Mol Cell Endocrinol 78:C113-C118, 1991.
neurons of the hypothalamo-pituitary-adrenocortical axis. J Neurosci 131. Labrie F, Belanger A, Cusan L, Gomez JL, Candas B. Marked decline
9: 3072-3082, 1989. in serum concentrations of adrenal C19 sex steroid precursors and con-
105. Herman JP, Seroogy K. Hypothalamic-pituitary-adrenal axis glucocor- jugated androgen metabolites during aging. J Clin Endocrinol Metab
ticoids and neurologic disease. Neurol Clin 24: 461-481, 2006. 82: 2396-2402, 1997.
106. Heuser I, Deuschle W, Weber A, Kniest A, Ziegler C, Weber B, Colla 132. Labrie F, Belanger A, Simard J, Van Luu-The, Labrie C. DHEA and
M. The role of mineralocorticoid receptors in the circadian activity peripheral androgen and estrogen formation: Intracrinology. Ann N Y
of the human hypothalamus-pituitary-adrenal system: Effect of age. Acad Sci 774: 16-28, 1995.
Neurobiol Aging 21: 585-589, 2000. 133. Labrie F, Dupont A, Belanger A. Complete androgen blockade for
107. Higuchi K, Ogo A, Maki T, Haji M, Takayanagi R, Ohashi M, Nawata the treatment of prostate cancer. Important Adv Oncol 1985: 193-217,
H, Kato K, Ibayashi H. Evidence for age-related change in plasma 1985.
19-hydroxyandrostenedione. Endocrinol Jpn 36: 881-885, 1989. 134. Lai M, Horsburgh K, Bae SE, Carter RN, Stenvers DJ, Fowler JH,
108. Hill MN, Tasker JG. Endocannabinoid signaling glucocorticoid- Yau CE, Gomez-Sanchez CE, Holmes MC, Kenyon CJ, Seckl JR,
mediated negative feedback and regulation of the HPA axis. Neuro- MacLeod MR. Forebrain mineralocorticoid receptor overexpression
science 204: 5-16, 2012. enhances memory, reduces anxiety and attenuates neuronal loss in cere-
109. Holanda CM, Guerra RO, Nobrega PV, Costa HF, Piuvezam MR, bral ischemia. Eur J Neurosci 25: 1832-1842, 2007.
Maciel AC. Salivary cortisol and frailty syndrome in elderly residents of 135. Lee SY, Hwang YK, Yun HS, Han JS. Decreased levels of nuclear
long-stay institutions: A cross-sectional study. Arch Gerontol Geriatr glucocorticoid receptor protein in the hippocampus of aged Long-Evans
54:e146-e151, 2012. rats with cognitive impairment. Brain Res 1478: 48-54, 2012.
110. Holta H, Achida S. Aging of the autonomic nervous system and possible 136. Leng SX, Cappola AR, Andersen RE, Blackman MR, Koenig K, Blair
improvements in autonomic activity using somatic afferent stimulation. M, Walston JD. Serum levels of insulin-like growth factor-1 (IFG-1)
Geriatr Gerontol Int 10(Suppl S1): S127-S136, 2010. and dehydroepiandrosterone sulfate (DHEA-S), and their relationships
111. Issa AM, Rowe W, Gauthier S, Meaney MJ. Hypothalamic-pituitary- with serum interleukin-6, in the geriatric syndrome of frailty. Aging
adrenal activity in aged, cognitively impaired and cognitively unim- Clin Exp Res 16: 153-157, 2004.
paired rats. J Neurosci 10: 3247-3254, 1990. 137. Luger A, Deuster PA, Kyle SB, Gallucci WT, Montgomery LC, Gold
112. Ito U, Ohno K, Suganuma Y, Suzuki K, Inaba Y. Effect of steroid PW, Loriaux DL, Chrousos GP. Acute hypothalamic-pituitary-adrenal
on ischemic brain edema. Analysis of cytotoxic and vasogenic edema responses to the stress of treadmill exercise: Physiologic adaptations to
occurring during ischemia and after restoration of blood flow. Stroke physical training. N Engl J Med 316: 1309-1315, 1987.
11: 166-172, 1980. 138. Lundblad JR, Roberts JL. Regulation of proopiomelanocortin gene
113. Jacobson L, Sapolsky R. The role of hippocampus in feedback regula- expression in pituitary. Endocr Rev 9: 135-158, 1988.
tion of the hypothalamic-pituitary-adrenocortical axis. Endocr Rev 12: 139. Lupien SJ, de Leon M, de Santi S, Convit A, Tarshish C, Nair NP,
118-134, 1991. Thakur M, McEwan BS, Hauger RL, Meaney MJ. Cortisol levels during
114. Jöels M, Krugers H, Karst H. Stress-induced changes in hippocampal human aging predict hippocampal atrophy and memory deficits. Nat
function. Prog Brain Res 167: 3-15, 2008. Neurosci 1: 69-73, 1998.
115. Joels M, Karst H, de Rijk RH, de Kloet ER. The coming out of the 140. Lupien SJ, Fiocco A, Wan N, Maheu F, Lord C, Schramek T, Tu
brain mineralocorticoid receptor. Trends Neurosci 31: 1-7, 2008. MT. Stress hormones and human memory function across the lifespan.
116. Joels M, Karst H, Krugers HJ,Lucassen PJ. Chronic stress: Implications Psychoneuroendocrinology 30: 225-242, 2005.
for neuronal morphology, function and neurogenesis. Front Neuroen- 141. Lupien S, Lecours AR, Schwartz G, Sharma S, Hauger RL, Meaney MJ,
docrinol 28: 72-96, 2007. Nair NP. Longitudinal study of basal cortisol levels in healthy elderly
117. Joels M, Puz Z, Wiegert O, Oitzl MS, Krugers HJ. Learning under subjects: Evidence for subgroups. Neurobiol Aging 17: 95-105, 1996.
stress: How does it work? Trends Cogn Sci 10: 152-158, 2006. 142. Lupien SJ, Nair NP, Briere S, Maheu F, Tu MT, Lemay M, McEwan
118. Jun SS, Chen Z, Pace MC, Shaul PW. Glucocorticoids downregulate BS, Meaney MJ. Increased cortisol levels and impaired cognition in
cyclooxygenase-1 gene expression and prostacyclin synthesis in fetal human aging: Implication for depression and dementia in later life. Rev
pulmonary artery endothelium. Circ Res 84: 193-200, 1999. Neurosci 10: 117-139, 1999.
119. Jung FF, Kennefick TM, Ingelfinger JR, Vora JP, Anderson S. Down- 143. MacLullich AM, Ferguson KJ, Wardlaw JM, Starr JM, Deary IS,
regulation of the intrarenal renin-angiotensin system in the aging rat. Seckl JR. Smaller left anterior cingulate cortex volumes are associ-
J Am Soc Nephrol 5: 1573-1580, 1995. ated with impaired hypothalamic-pituitary-adrenal axis regulation in
120. Kalmijn S, Launer LJ, Stolk Rp, de Jong FH, Pols HA, Hofman A, healthy elderly men. J Clin Endocrinol Metab 91: 1591-1594, 2006.
Breteler MM, Lamberts SW. A prospective study on cortisol, dehy- 144. MacPherson A, Dinkel K, Sapolsky R. Glucocortocoids worsen
droepiandrosterone sulfate, and cognitive function in the elderly. J Clin excitotoxin-induced expression of pro-inflammatory cytokines in hip-
Endocrinol Metab 83: 3487-3492, 1998. pocampal cultures. Exp Neurol 194: 376-383, 2007.
121. Kant GF, Meyerhoff JL, Jarrard LE. Biochemical indices of reactivity 145. Maeda K, Tanimoto K, Terada T, Shintani T, Kakiqi T. Elevated urinary
and habituation in rats with hippocampal lesions. Pharmacol Biochem free cortisol in patients with dementia. Neurobiol Aging 12: 161-163,
Behav 20: 793-797, 1984. 1991.
122. Karst H, Berger S, Turiault M, Tronche F, Schutz G, Joels M. Miner- 146. Maki T. Age-related changes in secretion of adrenocortical steroid
alocorticoid receptors are indispensable for nongenomic modulation of hormones in normal healthy men. [Japanese] Nihon Naibunpi Gakkai
hippocampalglutamate transmission by corticosterone. Proc Natl Acad Zasshi 62: 672-682, 1986.
Sci 102: 19204-19207, 2005. 147. Martignoni E, Costa A, Sinforiani E, Liuzzi A, Chiodini P, Mauri
123. Kasckow J, Xiao C, Herman JP. Glial glucocorticoid receptors in aged M, Bono G, Nappi G. The brain as a target for adrenocortical steroids:
Fisher 344 (F344) and F344/Brown Norway rats. Exp Gerontol 44: Cognitive implications. Psychoneuroendocrinology 17: 343-354, 1992.
335-343, 2009. 148. Marissal-Arvy N, Lombes M, Petterson J, Moisan MP, Mormede P.
124. Keller-Wood ME, Dallman MF. Cortocosteroid inhibition of ACTH Gain of function mutation in the mineralocorticoid receptor of the
secretion. Endocr Rev 5: 1-24, 1984. Brown Norway rat. J Biol Chem 279: 39232-39239, 2004.
125. Kinsella K, Velkoff VA. U.S. Census Bureau, Series P95/01-1. An 149. Mayo W, Lemaire V, Malaterre J, Rodriguez JJ, Cayre M, Stewart
Aging World: 2001. U.S. Govt Printing Office; Washington, DC. MG, Kharouby M, Rougon G, Le Moal M, Piazza PV, Abrous DN.

Volume 4, October 2014 1507


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

Pregnenolone sulfate enhances neurogenesis and PSA-NCAM in young 174. Muller MB, Lucassen PJ, Yassouridis A, Hoogendijk WJG, Holsboer
and aged hippocampus. Neurobiol Aging 26: 103-114, 2005. F, Swaab DF. Neither major depression no glucocorticoid treatment
150. McEwen BS. Mood disorders and allostatic load. Biol Psychiatry 54: affects the cellular integrity of the human hippocampus. Eur J Neurosci
200-207, 2003. 14: 1603-1612, 2001.
151. McEwen BS, de Kloet ER, Rostene W. Adrenal steroid receptors and 175. Munoz CD, Lepsch LB, Kawamoto EM, Malta MB, Lima Lde S, Avel-
actions in the nervous system. Physiol Rev 66: 1121-1188, 1986. lar MC, Sapolsky RM, Scarone C. Chronic unpredictable stress exacer-
152. McEwen BS, Wingfield JC. The concept of allostasis in biology and bates lipopolysaccharide-induced activation of nuclear factor kappaB
biomedicine. Horm Behav 43: 2-15, 2003. in the frontal cortex and hippocampus via glucocorticoid secretion.
153. McGaugh JL. The amygdala modulates the consolidation of memories J Neurosci 26: 3813-3820, 2006.
of emotionally arousing experiences. Annu Rev Neurosci 27: 1-28, 176. Munoz CD, Sorrells SF, Caso JR, Scavone RM. Glucocorticoids
2004. exercerbate lipopolysaccharide-induced signaling in the frontal cortex
154. Meaney MJ, Aitken DH. Dexamethasone binding in rat frontal cortex. and hippocampus in a dose-dependent manner. J Neurosci 30: 13690-
Brain Res 328: 176-180, 1985. 13698, 2010.
155. Meaney MJ, Aitken DH, Sharma S, Viau V. Basal ACTH, corticos- 177. Murialdo G, Barreca A, Nobili F, Rollero A, Timossi G, Gianelli MV,
terone and corticosterone-binding globulin levels over the diurnal cycle, Copello F, Rodriguez G, Polleri A. Dexamethasone effects on cortisol
and age-related changes in hippocampal type I and type II corticosteroid secretion in Alzheimer’s disease: Some clinical and hormonal features
receptor binding capacity in young and aged, handled and nonhandled in suppressor and nonsuppressor patients. J Endocrinol Invest 23: 178-
rats. Neuroendocrinology 55: 204-213, 1992. 186, 2000.
156. Meaney MJ, Mitchell JB, Aitken DH, Bhatnagar D, Bodnoff SR, Iny LJ, 178. Murphy EK, Spencer RL, Sipe KJ, Herman JP. Decrements in nuclear
Sarrieau A. The effects of neonatal handling on the development of the glucocorticoid receptor (GR) protein levels and DNA binding in aged
adrenal cortical response to stress: Implications for the neuropathology rate hippocampus. Endocrinology 143: 1362-1370, 2002.
and cognitive deficits in later life. Psychoneuroendocrinology 16: 85- 179. Nicolson N, Storms C, Ponds R, Sulon J. Salivary cortisol levels and
103, 1991. stress reactivity in human aging. Gerontol A Biol Sci Med Sci 52: M68-
157. Meaney MJ, Szyf M, Seckl JR. Epigenetic mechanisms of perinatal M75, 1997.
programming of hypothalamic-pituitary-adrenal function and health. 180. Noordam R, Jansen SW, Akintola AA, Oei NY, Maier AB, Pijl H, Slag-
Trends Mol Med 13: 269-277, 2007. boom PE, Westendorp RG, van der Goud J, de Craen AJ, van Heemst
158. Michaud K, Forget H, Cohen H. Chronic glucocorticoid hypersecretion D; Leiden Longevity Study Group. Familial Longevity Is Marked by
in Cushing’s syndrome exacerbates cognitive aging. Brain Cogn 71: Lower Diurnal Salivary Cortisol Levels: The Leiden Longevity Study.
1-8, 2009. PLoS ONE 7: e31166, 2012.
159. Michelson O, Chrousos GP, Gold PW. Type I glucocorticoid recep- 181. Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin
tor blockade does not affect baseline or ovine corticotropin-releasing- AM. Prenatal exposure to maternal depression, neonatal methylation of
hormone-stimulated adrenocorticotropic hormone and cortisol secre- human glucocorticoid receptor gene (NR3C1) and infant cortisol stress
tion. NeuroImmunoModulation 1: 274-277, 1994. responses. Epigenetics 3: 97-106, 2008.
160. Mikics E, Kruk MR, Haller J. Genomic and non-genomic effects of 182. O’Brien JT, Desmond P, Ames D, Schweitzer I, tuckwell V, Tress B. The
glucocorticoids on aggressive behavior in male rats. Psychoneuroen- differentiation of depression from dementia by temporal lobe magnetic
docrinology 29: 618-635, 2004. resonance imaging. Psychol Med 24: 633-640, 1994.
161. Milcu SM, Bogdan C, Nicolau GY, Cristea A. Cortisol circadian rhythm 183. Oitzl MS, Champagne DL, van der Veen R, de Kloet ER. Brain devel-
in 70-100-year-old subjects. Endocrinologie 16: 29-39, 1978. opment under stress: Hypotheses of glucocorticoid actions revisited.
162. Miller BS, Ignatoski KM, Daignault S, Lindland C, Gauger PG, Doherty Neurosci Biobehav Rev 34: 853-866, 2010.
GM, Wang SC; University of Michigan Analytical Morphomics Group. 184. Oitzl MS, Fluttert M, de Kloet ER. The effect of cortocosterone on reac-
A quantitative tool to assess degree of sarcopenia objectively in patients tivity to spatial novelty is mediated by central mineralocorticosteroid
with hypercortisolism. Surgery 150: 1178-1185, 2011. receptors. Eur J Neurosci 6: 1072-1079, 1994.
163. Mizoguchi K, Ikeda R, Shoji H, Tanaka Y, Maruyama W, Tabira T. 185. Oitzl MS, Reichardt HM, Joels M, de Kloet ER. Point mutation in the
Aging attenuates glucocorticoid negative feedback in rat brain. Neuro- mouse glucocorticoid receptor preventing DNA binding impairs spatial
science 159: 259-270, 2009. memory. Proc Natl Acad Sci USA 98: 12790-12795, 2001.
164. Mizoguchi K, Ishige A, Aburada M, Tabira T. Chronic stress attenuates 186. Orth DN. Corticotropin-releasing hormone in humans. Endocr Rev 13:
glucocorticoid negative feedback: Involvement of the prefrontal cortex 164-191, 1992.
and hippocampus. Neuroscience 119: 887-897, 2003. 187. Otte C, Jahn H, Yassourids A, Arlt J, Stober N, Maass P, Wiedemann
165. Morimoto S, Takeda R, Uchida K, Miyamori I, Miyamoto M, Mimou K, Kellner M. The mineralcorticoid receptor agonist, fludrocortisone,
N, Kigoshi T. Reduced aldosterone secretory response to acute ACTH inhibits pituitary-adrenal activity in humans after pre-treatment with
stimulation in sodium-restricted elderly subjects. J Am Geriatr Soc 28: metyrapone. Life Sci 73: 1835-1845, 2003.
361-366, 1980. 188. Otte C, Moritz A, Yassouridis A, Koop M, Madrischewski AM, Wiede-
166. Morley JE. Developing novel therapeutic approaches to frailty. Curr mann K, Kellner M. Blockade of the mineralocorticoid receptor in
Pharm Des 15: 3384-3395, 2009. healthy men: Effects on experimentally induced panic symptoms,
167. Morley JE, Kaiser F, Raum WJ, Perry HM III, Flood JF, Jensen J, Silver stress hormones, and cognition. Neuropsychopharmacology 32: 232-
AJ, Roberts E. Potentially predictive and manipulable blood serum 238, 2007.
correlates of aging in the healthy human male: Progressive decreases in 189. Otte C, Yassouridis A, Jahn H, Maass P, Stober N, Wiedemann
bioavailable testosterone, dehydroepiandrosterone sulfate, and the ratio K, Kellner M. Mineralocorticoid receptor-mediated inhibition of the
of insulin-like growth factor 1 to growth hormone. Proc Natl Acad Sci hypothalamic-pituitary-adrenal axis in aged humans. J Gerontol A Biol
U S A 94: 7537-7542, 1997. Sci Med Sci 58: 900-905, 2003.
168. Morley JE, Malmstrom TK, Miller DK. A simple frailty questionnaire 190. Parker L, Gral T, Perrigo V, Skowksy R. Decreased adrenal andro-
(FRAIL) predicts outcomes in middle aged African Americans. J Nutr gen sensitivity to ACTH during aging. Metabolism 30: 601-604,
Health Aging 16: 601-608, 2012. 1981.
169. Morley JE, Vellas B, van Kan GA, Anker SD, Bauer JM, Bernabei 191. Pavlov EP, Harman SM, Chrousos GP, Loriaux OL, Blackman
R, Cesari M, Chumlea WC, Doehner W, Evans J, Fried LP, Gural- MR. Responses of plasma adrenocorticotropin, cortisol, and dehy-
nik JM, Katz PR, Malmstrom TK, McCarter RJ, Gutierrez-Robledo droepiandrosterone to ovine corticotropin-releasing hormone in healthy
LM, Rockwood K, von Haehling S, Vandewoude MF, Walston J. aging men. J Clin Endocrinol Metab 62: 767-772, 1986.
Frailty consensus: A call to action. J Am Med Dir Assoc 14: 392-397, 192. Peeters GM, van Schoor NM, Visser M, Knol DL, Eekhoff EM,
2013. de Ronde W, Lips P. Relationship between cortisol and physi-
170. Morsink MC, Joels M, Sarabdjitsingh RA, Meijer OC, de Kloet ER, cal performance in older persons. Clin Endocrinol 67: 398-406,
Datson NA. The dynamic pattern of glucocorticoid receptor-mediated 2007.
transcriptional responses in neuronal PC12 cells. J Neurochem 99: 193. Phelps EA, LeDoux JE. Contributions of the amygdala to emotion
1282-1298, 2006. processing: From animal models to human behavior. Neuron 48: 175-
171. Morsink MC, Steenbergen PJ, Vos JB, Karst H, Joels M, de Kloet ER, 187, 2005.
Datson NA. Acute activation of hippocampal glucocorticoid receptors 194. Pimenta E, Calhoun DA. Resistant hypertension and aldosteronism.
results in different waves of gene expression throughout time. J Neu- Curr Hypertens Rep 9: 353-359, 2007.
roendocrinol 18: 239-252, 2006. 195. Potter CL, Goodfriend Tl. Aldosterone production and hormone respon-
172. Mountjoy KG, Robbins LS, Mortrud MT, Cone RD. The cloning of a siveness in adrenal glomerulosa cells from cows of different age. Geron-
family of genes that encode the melanocortin receptors. Science 257: tology 33: 77-86, 1987.
1248-1251, 1992. 196. Prinz P, Bailey S, Moe K, Wilkinson C, Scanlan J. Urinary free corti-
173. Mulchahey JJ, Kasckow JW, Plotsky PM, Hauger RL. Steroidal reg- sol and sleep under baseline and stressed conditions in healthy senior
ulation of portal arginine-vasopressin levels in aged Fischer 344 rats. women: Effects of estrogen replacement therapy. J Sleep Res 10: 19-26,
Brain Res 822: 243-245, 1999. 2001.

1508 Volume 4, October 2014


Comprehensive Physiology Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging

197. Prinz PN, Bailey SL, Woods DL. Sleep impairments in healthy seniors: 219. Sapolsky RM, Romero LM, Munck AU. How do glucocorticoids influ-
Roles of stress, cortisol, and interleukin-1 beta. Chronobiol Int 17: ence stress responses? Integrating permissive, suppressive, stimulatory,
391-404, 2000. and preparative actions. Endocr Rev 21: 55-89, 2000.
198. Purnell JQ, Brandon DD, Isabelle LM, Loriaux DL, Samuels MH. Asso- 220. Scherrer LC, Dalman FC, Massa E, Meshinchi S, Pratt WB. Struc-
ciation of 24-hour cortisol production rates, cortisol-binding globulin, tural and functional reconstitution of the glucocorticoid receptor-hsp90
and plasma-free cortisol levels with body composition, leptin levels, and complex. J Biol Chem, 265: 21397-21400, 1990.
aging in adult men and women. J Clin Endocrinol Metab 89: 281-287, 221. Seals DR, Esler MD. Human aging and the sympathoadrenal system.
2004. J Physiol 528: 407-417, 2000.
199. Quirk GJ, Beer JS. Prefrontal involvement in the regulation of emotion: 222. Seckl JR, Holmes MC. Mechanisms of disease: Glucocorticoids, their
Convergence of rat and human studies. Curr Opin Neurobiol 16: 723- placental metabolism and fetal ‘programming’ of adult pathophysiol-
727, 2006. ogy. Nat Clin Pract Endocrinol Metab 3: 479-488, 2007.
200. Radke KJ. Age-related impairment of K(+)- and ACTH-stimulated 223. Seeman TE, McEwen BS, Singer Bh, Albert MS, Rowe JW. Increase
aldosterone secretion by rat adrenal capsules. Am J Physiol 264(1 Pt 1): in urinary cortisol excretion and memory declines: MacArthur studies
E82-E89, 1993. of successful aging. J Clin Endocrinol Metab 82: 2458-2465, 1997.
201. Rao MY, Rao TS, Narayanaswamy RK. Study of hypothalamo pituitary 224. Seeman TE, Robbins RJ. Aging and hypothalamic-pituitary-adrenal
adrenal axis in frail elderly subjects. J Assoc Physicians India 60: 31-34, response to challenge in humans. Endocr Rev 15: 233-260, 1994.
2012. 225. Serio M, Piolanti Cappelli G, De Magistris L, Ricci F, Anzalone M,
202. Reichardt HM, Kaestner J, Tuckermann J, Kretz O, Wessely O, Bock R, Giusti G. The miscible pool and turnover rate of cortisol in the aged,
Gass P, Schmid W, Herrlich P, Angel P, Schutz G. DNA binding of the and variations in relation to time of day. Exp Gerontol 4: 95-101,
glucocorticoid receptor is not essential for survival. Cell 93: 531-541, 1969.
1998. 226. Silverman MN, Sternberg EM. Glucocorticoid regulation of inflam-
203. Reul JMHM, de Kloet ER. Two receptor systems for corticosterone in mation and its functional correlates: From HPA axis to glucocorticoid
rat brain: Microdistribution and differential occupation. Endocrinology receptor dysfunction. Ann N Y Acad Sci 1261: 55-63, 2012.
117: 2505-2511, 1985. 227. Simpson ER, Waterman MR. Regulation of the synthesis of steroido-
204. Reul JM, Rothuizen J, de Kloet ER. Age-related changes in the dog genic anzymes in adrenal cortical cells by ACTH. Annu Rev Physiol
hypothalamic-pituitary-adrenocortical system: Neuroendocrine activ- 50: 427-440, 1988.
ity and corticosteroid receptors. J Steroid Biochem Mol Biol 40: 63-69, 228. Smith AI, Funder JW. Proopiomelanocortin processing in the pituitary,
1991. central nervous system, and peripheral tissues. Endocr Rev 9: 159-179,
205. Ribarac-Stepic N, Vulovic M, Koricanac G, Isenovic E. Basal and glu- 1988.
cocorticoid induced changes of hepatic glucocorticoid receptor during 229. Sorrells SF, Caso JR, Munhoz CD, Hu CK, Tran KV, Miguel ZD, Chien
aging: Relation to activities of tyrosine aminotransferase and trypto- BY, Sapolsky RM. Glucocorticoid signaling in myeloid cells worsens
phan oxygenase. Biogerontology 6: 113-131, 2005. acute CNS injury and inflammation. J Neurosci 33: 7877-3789, 2013.
206. Ridder S, Chourbaji S, Hellweg R, Urani A, Zacher C, Schmid W, Zink 230. Sousa N, Madeira MD, Paula-Barbosa MM. Effects of corticosterone
M, Hortnagl H, Flor H, Henn FA, Schutz G, Gass P. Mice with geneti- treatment and rehabilitation on the hippocampal formation of neonatal
cally altered glucocorticoid receptor expression show altered sensitiv- and adult rats. An unbiased stereological study. Brain Res 794: 199-210,
ity for stress-induced depressive reactions. J Neurosci 25: 6245-6250, 1998.
2005. 231. Starkman MN, Gebarski SS, Berent S, Schteingart DE. Hippocampal
207. Roberts I, Yates D, Sandercock P, Farrell B, Wasserberg J, Lomas G, formation volume, memory dysfunction, and cortisol levels in patients
Cottingham R, Svoboda P, Brayley N, Mazairac G, Laloë V, Munoz- with Cushing’s syndrome. Biol Psychiatry 32: 756-765, 1992.
Sanchez A, Arango M, Hartzenberg B, Khamis H, Yutthakasemsunt 232. Starkman MN, Giordani B, Gebarski SS, Berent S, Schork MA,
S, Komolafe E, Olldashi F, Yadav Y, Murillo-Cabezas F, Shakur H, Schteingart DE. Decrease in cortisol reverses human hippocampal atro-
Edwards P, CRASH Trial collaborators. Effect of intravenous corticos- phy following treatment of Cushing’s disease. Biol Psychiatry 46: 1595-
teroids on death within 14 days in 10008 adults with clinically signifi- 1602, 1999.
cant head injury (MRC CRASH trial: Randomized placebo-controlled 233. Stocco DM, Clark BJ. Regulation of the acute production of steroids in
trial). Lancet 364: 1321-1328, 2004. steroidogenic cells. Endocr Rev 17: 221-244, 1996.
208. Rolland Y, Czerwinski S, Abellan van Kan G, Morley JE, Cesari M, 234. Swanwick GR, Kirby M, Bruce I, Buggy F, Coen RF, Coakley
Onder G, Woo J, Baumgartner R, Pillard F, Boirie Y, Chumlea WM, D, Lawlor BA. Hypothalamic-pituitary-adrenal axis dysfunction in
Vallas B. Sarcopenia: Its assessment, etiology, pathogenesis, conse- Alzheimer’s disease: Lack of association between longitudinal and
quences and future perspectives. J Nutr Health Aging 12: 433-450, cross-sectional findings. The Am J Psychiatry 155: 286-289, 1998.
2008. 235. Takeda Y, Miyamori I, Yoneda T, Iki K, Hatakeyama H, Takeda R.
209. Rolland YM, Perry HM III, Patrick P, Banks WA, Morley JE. Loss Effect of aging on urinary excretion of 18-hydroxycortisol. J Steroid
of appendicular muscle mass and loss of muscle strength in young Biochem Mol Biol 50: 167-168, 1994.
postmenopausal women. J Gerontol A Biol Sci Med Sci 62:B330-B335, 236. Takeda Y, Miyamori I, Yoneda T, Iki K, Takeda R. Effect
2007. of aging on urinary excretion of 19-noraldosterone and 18,19-
210. Ros-Bernal F, Hunot S, Herrero MJ, Parnadeau S, Corvol JC, Lu L, dihydroxycorticosterone. J Steroid Biochem Mol Biol 52: 383-386,
Alvarez-Fischer D, Carrillo-de Sauvage MA, Saurini F, Coussieu C, 1995.
Kinugawa K, Prigent A, Höglinger G, Hamon M, Tronche F, Hirsch 237. Tasker JG, Di S, Malcher-Lopes R. Minireview: Rapid glucocorti-
EC, Vyas S. Microglial glucocorticoid receptors play a pivotal role in coid signaling via membrane-associated receptors. Endocrinology 147:
regulating dopaminergic neurodegeneration in parkinsonism. Proc Natl 5549-5556, 2006.
Acad Sci U S A 108: 6632-6637, 2011. 238. Taylor AL, Fishman LM. Cortocotropin-releasing hormone. N Engl
211. Rozeboom AM, Akil H, Seasholtz AF. Mineralocorticoid receptor over- J Med 319: 213-222, 1988.
expression in forebrain decreases anxiety-like behavior and alters the 239. Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, Orbau PC, Bock
stress response in mice. Proc Natl Acad Sci U S A 104: 4688-4693, R, Klein R, Schütz G. Disruption of the glucocorticoid receptor gene in
2007. the nervous system results in reduced anxiety. Nat Genet 23: 99-103,
212. Rowe JW, Troen BR. Sympathetic nervous system and aging in man. 1999.
Endocr Rev 1: 167-179, 1980. 240. Tyler FH, Eik-Nes K, Sandberg AA, Florentin AA, Samuels LT.
213. Sanchez MM, Young LJ, Plotsky PPM, Insel TR. Distribution of corti- Adrenocortical capacity and the metabolism of cortisol in elderly
costeroid receptors in the rhesus brain: Relative absence of glucocorti- patients. J Am Geriatr Soc 3: 79-84, 1955.
coid receptors in the hippocampal formation. J Neurosci 20: 4657-4668, 241. Ulrich-Lai YM, Herman JP. Neural regulation of endocrine and auto-
2000. nomic stress responses. Nat Rev Neurosci 10: 397-409, 2009.
214. Sanders MJ, Siltgen BJ, Fanselow MS. The place of the hippocampus 242. Valenti G, Denti L, Maggio M, Ceda G, Volpato S, Bandinelli S,
in fear conditioning. Eur J Pharmacol 463: 217-223, 2003. Ceresini G, Cappola A, Guralnik JM, Ferrucci L. Effect of DHEAS and
215. Sandi C, Venero C, Guaza C. Novelty-related rapid locomotor effects skeletal muscle over the life span: The InCHIANTI study. J Gerontol
of corticosterone in rats. Eur J Neurosci 8: 794-800, 1996. A Biol Sci Med Sci 59: 466-472, 2004.
216. Sapolsky RM, Krey LC, McEwen BS. Glucocorticoid-sensitive hip- 243. Van Cauter E, Blackman JD, Roland D, Spire JP, Refetoff S, Polonsky
pocampal neurons are involved in terminating the adrenocortical stress KS. Modulation of glucose regulation and insulin secretion by circadian
response. Proc Natl Acad Sci U S A 81: 6174-6177, 1984. rhythmicity and sleep. J Clin Invest 88: 934-942, 1991.
217. Sapolsky RM, Krey LC, McEwen BS. Prolonged glucocorticoid expo- 244. Van Cauter E, Leproult R, Kupfer DJ. Effects of gender and age on the
sure reduces hippocampal neuron number: Implications for aging. levels and circadian rhythmicity of plasma cortisol. J Clin Endocrinol
J Neurosci 5: 1222-1227, 1985. Metab 81: 2468-2473, 1996.
218. Sapolsky RM, Krey LC, McEwen BS. The neuroendocrinology of stress 245. van Eekelen JA, Oitzl MS, de Kloet ER. Adrenocortical hyporespon-
and aging: The glucocorticoid cascade hypothesis. Endocr Rev 7: 284- siveness and glucocorticoid feedback resistance in old male brown
301, 1986. Norway rats. J Gerontol A Biol Sci Med Sci 50:B83-B89, 1995.

Volume 4, October 2014 1509


Hypothalamic-Pituitary-Adrenal (HPA) Axis and Aging Comprehensive Physiology

246. van Eekelen JA, Rots NY, Sutanto W, de Kloet ER. The effect of 255. Wellhoener P, Born J, Fehm HL, Dodt C. Elevated resting and exercise-
aging on stress responsiveness and central corticosteroid receptors in induced cortisol levels after mineralocorticoid receptor blockade with
the brown Norway rat. Neurobiol Aging 13: 159-170, 1992. canrenoate in healthy humans. J Clin Endocrinol Metab 89: 5048-5052,
247. Varadhan R, Walston J, Cappola AR, Carlson MC, Wand GS, Fried 2004.
LP. Higher levels and blunted diurnal variation of cortisol in frail older 256. West CD, Brown H, Simons EL, Carter DB, Kumagai LF, Englert E Jr.
women. J Gerontol A Biol Sci Med Sci 63: 190-195, 2008. Adrenocortical function and cortisol metabolism in older age. J Clin
248. Veith RC, Featherstone JA, Linares OA, Halter JB. Age differences Endocrinol Metab 21: 1197-1207, 1961.
in plasma norepinephrine kinetics in humans. J Gerontol 41: 319-324, 257. Wilkinson CW, Peskind ER, Raskind MA. Decreased hypothalamic
1986. pituitary-adrenal axis sensitivity to cortisol feedback inhibition in
249. Veldhuis JD, Sharma A, Roelfsema F. Age-dependent and gender- human aging. Neuroendocrinology 65: 79-90, 1997.
dependent regulation of hypothalamic-adrenocorticotropic-adrenal 258. Wolf OT, Convit A, Thorn E, de Leon MJ. Salivary cortisol day profiles
axis. Endocrinol Metab Clin N Am 42: 201-225, 2013. in elderly with mild cognitive impairment. Psychoneuroendocrinology
250. Vermeulen A, Deslypere JP, Schelfhout W, Verdonck L, Rubens 27: 777-789, 2002.
R. Adrenocortical function in old age: Response to acute adreno- 259. Wong YY, McCaul KA, Yeap BB, Hankey GJ, Flicker L. Low vitamin
corticotropin stimulation. J Clin Endocrinol Metab 54: 187-191, D status is an independent predictor of increased frailty and all-cause
1982. mortality in older men: The Health in Men Study. J Clin Endocrinol
251. Voznesensky M, Walsh S, Dauser D, Brindisi J, Kenny AM. The asso- Metab 98: 3821-3828, 2013.
ciation between dehydroepiandosterone and frailty in older men and 260. Woo J, Leung J, Morley JE. Comparison of frailty indicators based
women. Age Ageing 38: 401-406, 2009. on clinical phenotype and the multiple deficit approach in predicting
252. Waterman MR, Bishoff LJ. Cytochromes P450 12: Diversity of ACTH mortality and physical limitation. J Am Geriatr Soc 60: 1478-1486,
(cAMP)-dependent transcription of bovine steroid hydroxylase genes. 2012.
FASEB J 11: 419-427, 1997. 261. Yau JL, Noble J, Hibberd C, Rowe WB, Meaney MJ, Morris RG, Seckl
253. Waters DL, Qualis CR, Dorin RI, Veldhuis JD, Baumgartner RN. JR. Chronic treatment with the antidepressant amitriptyline prevents
Altered growth hormone, cortisol, and leptin secretion in healthy elderly impairments in water maze learning in aging rats. J Neurosci 22: 1436-
persons with sarcopenia and mixed body composition phenotypes. 1442, 2002.
J Gerontol A Biol Sci 63: 536-541, 2008. 262. Young EA, Lopez JF, Murphy-Weinberg V, Watson SJ, Akil H. The role
254. Weidmann P, De Myttenaere-Burztein S, Maxwell MH, de Lima J. of mineralcorticoid receptors in hypothalamo-pituitary-adrenocortical
Effect of aging on plasma renin and aldosterone in normal man. Kidney axis regulation in humans. J Clin Endocrinol Metab 83: 3339-3345,
Int 8: 325-333, 1975. 1998.

1510 Volume 4, October 2014

Potrebbero piacerti anche