Age-Related Changes of The Hypothalamic Pituitary Adrenal Axis: Pathophysiological Correlates

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

European Journal of Endocrinology (2001) 144 319±329 ISSN 0804-4643

INVITED REVIEW

Age-related changes of the hypothalamic±pituitary±adrenal


axis: pathophysiological correlates
E Ferrari, L Cravello, B Muzzoni, D Casarotti, M Paltro, S B Solerte, M Fioravanti, G Cuzzoni, B Pontiggia
and F Magri
Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
(Correspondence should be addressed to E Ferrari, Department of Internal Medicine and Medical Therapeutics, University of Pavia, Piazza Borromeo 2,
27100 Pavia, Italy; Email: ferrari@unipv.it)

Abstract
The aim of this review was to examine the evidence for age-related changes of the hypothalamic±
pituitary±adrenal (HPA) axis in both physiological and pathological aging, on the basis of the many
data in the literature, as well as of our personal findings.
A statistically significant circadian rhythmicity of serum cortisol was maintained in elderly
subjects, even if with a reduced amplitude of the 24 h fluctuations and a trend to an increase of the
serum levels in the evening and at night-time, in comparison with young controls. Furthermore, an
age-related impairment of HPA sensitivity to steroid feedback was present in elderly people.
The occurrence of senile dementia amplified the changes already present in physiological aging.
While the cortisol secretion was generally well maintained in aging, the adrenal production of
dehydroepiandrosterone and of its sulfate (DHEAS) exhibited an age-related decline. Therefore, the
cortisol/DHEAS molar ratio was significantly higher in elderly subjects and even more in demented
ones, than in young controls.
Due to the opposite effects of cortisol and DHEAS on the brain and particularly on the hippocampal
region, the imbalance between glucocorticoids and androgens occurring in physiological and even
more in pathological aging, may have adverse effects on the function of this region, whose key role in
learning and memory is well known.

European Journal of Endocrinology 144 319±329

neural cell impairment and the compensatory gliosis


Introduction are particularly evident at the level of the hypothala-
The hypothalamic±pituitary±adrenal (HPA) axis is an mus and of the hippocampal and limbic system. In
auto-regulating system with many modulatory particular the neuronal impairment of the suprachias-
mechanisms; due to such regulation, the circulating matic nucleus (3) may be responsible for alterations of
levels of glucocorticoids are highly variable, according the circadian temporal structure of the aged organism,
both to the spontaneous rhythmic fluctuations and to due to the role played by the same nucleus as central
the responses towards stressful conditions. Further- pacemaker of several circadian rhythms. Furthermore,
more, the HPA axis realizes a tight integration among the loss of neurons, especially affecting both the
the endocrine, nervous and immune systems, being its hypothalamus as well as the hippocampus and the
key point. limbic region, implies a reduction of the glucocorticoid
In this way, the HPA axis is one of the most receptors and hence an impaired regulation of the
important allostatic (or adaptive) systems; when active, adrenocortical secretion (4, 5). Indeed, the role played
it allows the organism to respond to a challenge, or in by the hippocampus, both in animals and in humans,
general to a stressful condition, but its inactivation is in maintaining the adrenocortical circadian rhythmi-
equally important, since the allostatic load can over- city (6), as well as in modulating the glucocorticoid
expose the organism to the mediators of neural, feedback control of adrenocorticotropin (ACTH) secre-
endocrine and immune stress, with different patho- tion (7±9) and the adrenocortical responses to stressful
physiological consequences (1, 2). conditions (10) is well known.
Significant morphological and functional changes Consequently, the hippocampal degenerative changes
affect with age the HPA axis at different levels both occurring in physiological and even more in pathological
in experimental animals and in human beings. The aging may be responsible for the impaired sensitivity to

q 2001 Society of the European Journal of Endocrinology Online version via http://www.eje.org
320 E Ferrari and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144

the steroid feedback and for a certain degree of had been hypothesized in Alzheimer's disease (AD)
hyperactivity of the HPA axis in elderly subjects (11). (20); in the following years much experimental work
However, the heterogeneity of the age-related HPA confirmed this suggestion (21, 22). Similar findings
patterns must be underlined; indeed, some individuals were found both in primates (23) and in humans; in
exhibit substantial changes while others maintain an particular, a significant relationship between the
HPA function similar to younger individuals. increasing cortisol levels over a period of 4 years and
Many factors might be potentially involved in the the impairment of explicit memory and selective
aging process and, in particular, in brain aging, with attention performances has been described in a long-
several metabolic, immune and neuroendocrine itudinal study (24).
changes playing a pathogenetic role. Due to the Furthermore, high cortisol levels with an impaired
primary role of the adrenal steroids in maintaining HPA sensitivity to steroid feedback are often described
the homeostasis and the adaptational organism in senile dementia, both of degenerative and vascular
responses to stress, it seems very interesting to study types (25±28).
the link between the corticosteroid secretion and the Besides, a positive relationship of the cortisol
limbic±hippocampal age-related changes. response to an i.v. glucose load with the degree of
In this review we particularly focus on the age- hippocampal atrophy and the severity of dementia has
related changes of the glucocorticoid and androgen been described in several demented patients (29);
secretion, by considering separately the two hormonal moreover the cortisol increase seems to be a very
patterns, due to the peculiar modifications affecting the sensitive marker of cognitive decline in AD patients
different adrenocortical zones in aging. For such (30).
studies we favored the evaluation of the circadian The conventional measures of total cortisol levels
fluctuations of blood corticosteroids, which may might underestimate the real degree of hypercortiso-
provide better information about the pathophysiology lism in AD patients, since a reduction of corticosteroid-
of the endocrine secretions in comparison with the binding globulin (CBG) levels has been found in about
pharmacological tests, which sometimes do not reflect 30% of the patients with this kind of dementia (31). In
the spontaneous hormonal release. Furthermore, the any case, the CBG plasma levels were similar in old
chronobiological evaluation of neuroendocrine func- subjects with high, low or normal cortisol values, and a
tions in elderly people may represent a clinically strong correlation between total and unbound cortisol
reliable tool to investigate the biology of the CNS and levels across subjects has been described (32).
particularly of the limbic±hypothalamic system, and A cross-sectional approach in studying the age-
hence reveal the pathophysiology of the cerebral aging. related changes of the HPA axis may offer only tentative
Together with physiological aging, senile dementia suggestions. However, information arising from serial
represents a clinical model very interesting for chron- blood samples collected every 4 h during the day and
obiological studies, either for the frequency of changes every 2 h during the night, may give a true picture of
of the central neurotransmitter pathways, or because of the hormonal plasma changes in groups of different age
the frequent occurrence of degenerative alterations of which may underline the interindividual variations.
the hypothalamic suprachiasmatic nucleus. Furthermore, the inclusion in the study of subjects
drug-free and without overt acute or chronic disease
allowed us to investigate the role of age and dementia in
Cortisol secretion and aging HPA changes, independently of individual health status.
Much evidence suggests that the primary defect in On these basis, we studied the circadian rhythm of
allostatic systems in aging might be a prolonged serum cortisol in a wide number of healthy old subjects
response to stressful conditions, due to the inability to …n ˆ 52; age 69±90 years), of old demented patients,
shut off the allostatic response after the end of the including both AD and multi-infarct dementia (vascu-
stress (12±16). Consequently the HPA axis would lar dementia (VD)) …n ˆ 35; age 69±90 years) and of
become less resilient with age in responding to young controls …n ˆ 22; age 19±43 years).The physio-
stimulations. logical cortisol circadian rhythmicity was maintained,
However, in spite of the subtle age-related changes of with a significant age-related increase of the nocturnal
HPA function, the circulating levels of glucocorticoid cortisol levels in physiological aging and even more in
show a relative constancy or even a trend toward an senile dementia, in comparison with young subjects
increase with aging. Indeed, the blood levels of both (Fig. 1).
cortisol and ACTH usually fall within the normal range Our data agree with other studies, showing the age-
in physiological aging, but a trend towards higher related increase of the cortisol nadir values, together
nocturnal levels is often reported (17±19). with the phase-advance of the onset of the circadian
The possible role of changes of the glucocorticoid cortisol rise (33). Due to these changes, a reduction of
signal in the age-related modifications of the CNS has the quiescent period of the cortisol secretion and hence
been often suggested. Indeed, since 1968, a link an increase of the cortisol signal throughout the 24 h
between high glucocorticoid levels and neuronal loss occur in elderly people in comparison with young one.

www.eje.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144 Aging and adrenal steroids 321

Figure 1 Serum DHEAS and cortisol circadian rhythm


(before and after DXM administration, 1 mg orally at
2300 h) in physiological and pathological brain aging and
in young controls (means^S.E.M.). Old subjects vs
young controls: a ˆ P , 0:05; b ˆ P , 0:01;
c ˆ P , 0:001: AD patients vs young controls:
d ˆ P , 0:05; e ˆ P , 0:01; f ˆ P , 0:001: AD patients
vs old subjects g ˆ P , 0:05; h ˆ P , 0:01;
i ˆ P , 0:001: L/D, light dark.

The relative increase of cortisol serum levels in performance of subjects (r ˆ 0:3580, P , 0:01),
the evening and at night-time is responsible for the the latter being evaluated by the Mini Mental
clear flattening of the cortisol circadian profile. State Examination (Table 1).
Indeed, the nocturnal increase of serum cortisol Since the crucial role played by the central cholin-
(calculated as difference between the values recorded ergic and serotoninergic pathways in the circadian
at 0800 and 0000 h) was significantly reduced in activation of the ACTH-secreting system at night-time
both physiological and pathological aging; moreover is well known (33, 34), the impairment of the cortisol
this reduction was negatively correlated to both the nocturnal increase in elderly subjects may be a marker
age (r ˆ 20:3718, P , 0:01) and the cognitive of a reduced activity of these pathways.

www.eje.org
322 E Ferrari and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144

Table 1 Cortisol nocturnal increase in Interesting results have been also reported (44) by
physiological and pathological brain aging and in the CRH stimulation under DXM suppression. Indeed,
young controls (means ^ S.E.M.).
in spite of the higher cortisol levels recorded in old
Young controls 16.10 ^ 1.39a,b subjects under basal conditions, the levels of both
Old subjects 9.26 ^ 0.88 ACTH and cortisol after DXM were not different in old
Demented patients 8.02 ^ 1.83 and in young subjects, whereas a significantly higher
a cortisol response to CRH occurred in old subjects.
P , 0:001 (Student's t-test) vs demented patients.
b
P , 0:001 vs old subjects. Some data in the literature (45, 46) suggest that
aging does not modify the pituitary and adrenal
responsiveness to exogenous CRH. However, other
A reduction of the melatonin signal, widely described studies concerning cortisol and ACTH response to
in aging, could also contribute to the changes of both CRH and vasopressin, suggest that the pituitary
cortisol and ACTH nocturnal increase in elderly gland might become more sensitive with aging to CRH
subjects. Indeed the pineal gland seems to play an alone or in association with vasopressin (12, 16, 47) in
inhibitory role on the HPA axis (35, 36), by modulating the presence of stressful conditions and of age-related
the adrenal sensitivity to the corticotropic stimulation diseases (48). In our experience (45), the response of
(37) and even by limiting the corticotropin response to cortisol and ACTH to a CRH stimulation test (ovine
stressful conditions (38). Our data concerning the CRH, 1 mg/kg i.v.) was not significantly different in 14
simultaneous evaluation of cortisol and melatonin healthy old women (69±83 years) and in 11 young
secretions in elderly subjects confirm the opposite women (20±40 years) (Table 3).
behavior of the two secretions in both physiological Age-related changes of the endogenous CRH or
and pathological aging (39, 40) (Table 2). vasopressin secretion may also occur (49). Indeed in
Age-related changes of the dynamic secretory AD patients the CRH immunoreactivity has been
pattern of HPA axis have also been described. In fact, described as increased in the hypothalamus and
the cortisol peak values recorded during insulin- decreased in other cerebral areas, suggesting the
induced hypoglycemia were generally higher in old existence of alterations of the glucocorticoid regulation
than in young subjects, in spite of the smaller glycemic of this peptide (49). Similar neuroendocrine changes
fall occurring in the elderly (41). Thus, the HPA axis in may also be suggested in physiological aging (50).
aged people seems to recover slowly from certain types The adrenal sensitivity to exogenous ACTH seems
of stress. not to be significantly modified during physiological
Higher cortisol levels in old than in young subjects aging and in the early stage of AD (51). In our
have been described during some pharmacological experience (52) the cortisol response to pulse injection
challenges, such as the dexamethasone (DXM) suppres- of a small dose of synthetic ACTH (Synacthen,
sion test, the stimulation by human or ovine cortico- 2500 ng, i.v., at 2030 h) was quite similar in young
tropin-releasing hormone (CRH) or by physostigmine and old healthy women; however, patients with overt
(12, 42), probably due to the age-related changes of the senile dementia exhibited a significant increase of both
HPA sensitivity to both exogenous and endogenous the duration and the amplitude of cortisol response, as
stimuli. However, it seems noteworthy that the basal well as a significant delay of the time to peak …73 ^
cortisol levels exhibit a high intraperson stability and 4:7 min†; when compared with old healthy subjects
that they are closely related to the feedback sensitivity …46 ^ 4:9 min† and with young controls …45 ^ 5 min†:
of the HPA axis towards a low dose (0.25 mg) of DXM Since the cortisol response to a pulse injection of a
(43), suggesting a genetic influence on the set point of small dose of Synacthen is similar to that induced by a
the HPA axis. stressor, it is possible to argue that also in humans the
Table 2 Relationship between melatonin and cortisol secretion in
adrenocortical responsiveness to stressful conditions is
physiological and pathological brain aging and in young controls increased and prolonged only when `senescence is
(means ^ S.E.M.). coupled with pathological conditions', in agreement
with previous studies on old animals (14).
Ratio between Ratio between An age-related decrease of the HPA sensitivity to the
cortisol and cortisol nadir
melatonin and melatonin
steroid feedback signal has been reported. In particular
circadian mesors peak both the rate of non-suppression and the residual
cortisol levels after DXM were higher in demented
Young controls 0.58 ^ 0.17a 0.05 ^ 0.01b patients than in age-matched controls, possibly in
Old subjects 0.93 ^ 0.14 0.34 ^ 0.09c relation to anxiety, depression, panic or restlessness
AD patients 1.33 ^ 0.20 0.58 ^ 0.11
(53). Since these clinical symptoms and the high
AD patients and young controls: age vs cortisol nadir/melatonin peak ratio cortisol levels may be modified by the selective
r ˆ 0:64; P , 0:001:
a
serotonin re-uptake blockers, it is possible to suggest
P , 0:001 vs AD patients.
b
P , 0:01 vs AD patients. that the enhanced activity of the HPA axis in senile
c
P , 0:001 vs AD patients. dementia may be related to a central neurotransmitter

www.eje.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144 Aging and adrenal steroids 323

Table 3 Plasma ACTH and cortisol response to CRH stimulation elderly subjects and especially in demented patients
test (ovine-CRH, 1 mg/kg i.v.) in young and old healthy women than in young controls; moreover, patients with senile
(means ^ S.E.M.).
dementia also exhibited a delay of the cortisol response
Old healthy Young healthy to DXM (67).
women women These findings underline the relevance of the `age'
…n ˆ 14; …n ˆ 11; factor in the pathogenesis of the impaired HPA
age 69±83 years) age 20±40 years) sensitivity towards the negative steroid feedback, and
ACTH (pg/ml)
the additional effect of the occurrence of senile
Baseline 27.1 ^ 3.7 19.4 ^ 1.5 dementia.
Delta (max.) 29.2 ^ 8.4 28.1 ^ 10 In conclusion, the changes of the cortisol secretory
Delta (%) 117.9 ^ 22.3 144.8 ^ 33.5 pattern observed in elderly subjects and especially in
Cortisol (mg/dl) patients with senile dementia (namely the flattening of
Baseline 15.7 ^ 1.3 9.0 ^ 1.1
Delta (max.) 12.6 ^ 2.4 8.4 ^ 1.5 the circadian profile and the increase of the nadir
Delta (%) 90.4 ^ 18.5 93.3 ^ 21.2 levels, together with the impaired sensitivity to the
steroid feedback) are reminiscent of the data described
in senescent animals or in animals with experimental
imbalance, probably responsible also for the impaired hippocampal lesions (68).
sensitivity to the steroid feedback (53, 54).
It is now well established that the glucocorticoid
feedback on the HPA axis includes at least three time Dehydroepiandrosterone sulfate
domains: fast (seconds or minutes), intermediate (DHEAS) and aging
(usually less than 10 h) and slow (within hours or Only in humans and a few primates do the adrenals
days) (55), and that for all of them the sites of action secrete dehydroepiandrosterone (DHEA) and its sulfate
are the hypothalamus and the hippocampus (56), two (DHEAS) (69, 70); in particular, the zona reticularis of
brain areas in which the corticosteroid receptors (both the adrenal cortex is the exclusive source for DHEA and
type I and type II) are widely represented (57). DHEAS, and the level of expression of 3b-hydroxyster-
In order to study the first phase of the negative oid dehydrogenase is the most important determinant
feedback, Boscaro et al. (9) recently evaluated in aged for their biosynthesis (71).
people and in young controls the ACTH and cortisol In contrast with the minimal changes of the cortisol
suppressibility by the infusion of hydrocortisone, which secretion occurring with age, the last decades of life are
differs from DXM in the affinity for corticosteroid characterized by a progressive reduction of the secre-
receptors subtypes (16, 58±61). In spite of a similar tion rate of DHEA and DHEAS, the main adrenal
cortisol increase in both groups of subjects, the old ones androgens.
exhibited a slight and not significant decrease of ACTH In fact, the blood levels of these steroids, very low in
levels during the first phase of the response to the first years of life, begin to rise at the adrenarche and
hydrocortisone infusion, in comparison with young progressively increase till the third decade; after the age
controls; on the contrary, a pronounced and significant of 30 the steroid levels declines at a rate of 1±2% per
decline of ACTH concentrations occurred thereafter in year, even if with marked interindividual differences,
old subjects. and by the age of 70±80 they correspond only to 20±
These findings suggest that the age-related changes 30% of the peak concentration occurring in young
of the HPA activity in aging may be related to several people (72±74). The DHEAS/DHEA ratio does not
factors, namely a decrease with aging of the brain change with aging.
corticosteroid receptor concentration (62), but also the In spite of the great interindividual variability, the
age-related differences in the cerebrospinal fluid (CSF) DHEAS levels show a low within-subjects changes;
steroid concentrations (63), probably secondary to therefore they may be considered as highly specific
changes of the cortisol clearance rate in CSF and/or individual markers (75).
of the blood±brain barrier (64, 65). Thus, also vascular The morphological correlates of the physiological
factors may play a pathogenetic role in the decline of age-related changes of DHEA and DHEAS secretion are
the HPA sensitivity to the steroid feedback. the modifications of the zona reticularis. Indeed, the
In physiological and pathological brain aging and in development of this zone of the adrenals begins at the
young controls, the study of the circadian rhythm of adrenarche and shows a clear width reduction during
serum cortisol, both before and after DXM administra- aging (76). The mechanisms responsible for this
tion (1 mg at 2300 h, orally) (Fig. 1), allowed us to reduction are still unknown; furthermore, a significant
detect a high percentage of `non-responders' (serum correlation between the DHEAS decline in aging and
cortisol levels above 5 mg/dl the morning following the morphological changes of the zona reticularis was
DXM (66), in healthy old subjects (about 30%) and in never found (77).
old demented patients (50%). Indeed, the mean cortisol However, the zona reticularis is particularly suscep-
values recorded after DXM were significantly higher in tible to the intra-adrenal gradient of autocrine and

www.eje.org
324 E Ferrari and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144

paracrine factors created by the blood supply, and to an important homeostatic control mechanism for the
vascular damages, such as multiple microhemorragic neuronal activity, by binding to the g-aminobutyric
events, with subsequent necrotic damage. Taken acid A (GABAA) receptor (88). In particular, DHEA
together, all these factors might explain the age-related and DHEAS, acting as allosteric antagonists of
decline and the interindividual changes in DHEAS GABAA, receptor, may enhance the neuronal and
secretion. glial survival and may improve learning and memory
In addition to the reduced secretion in basal capacities (89, 90). Indeed in experimental animals,
conditions, the DHEAS response to exogenous ACTH DHEA administration has a memory-enhancing effect
is significantly impaired in old subjects, whereas the and in vitro studies suggest a neurotropic effect on
cortisol response is generally maintained (78). Thus, a neurons and glial cells (91).
specific, but unexplained, defect of the desmolase Furthermore, the noradrenergic release of N-methyl-
activity of P450c17 cytochrome in the zona reticularis d-aspartate in the rat hippocampus may be enhanced
seems to occur with aging (79). by DHEA (92). This action, together with the proser-
Extra-adrenal factors may also affect the adrenal otoninergic activity of DHEAS, could be responsible for
androgen biosynthesis. For example, some proopio- the antidepressive effects, and for the improvement of
melanocortin-related peptides, and in particular the memory reported after DHEA administration in
joint peptide and beta-endorphin, could stimulate the humans, due to the key role played by noradrenaline
androgen secretion (80, 81), although some authors on this function (93±95).
did not confirm such action (82, 83). The possible role In rat primary hippocampal neurons, the addition of
of a specific pituitary adrenal androgen-stimulating DHEAS, but not of DHEA, was able to enhance the
factor, identified by Parker et al. (81), is still debated. neuronal resistance to stress relevant for both acute
Furthermore, a possible influence of insulin in the and chronic neurodegenerative conditions, by the
age-related decline of DHEAS has been suggested, due activation of a kB-binding transcription factor (96)
to the inhibitory activity of insulin on DHEAS secretion, which is involved in the neuroprotection against
and the trend to increase of insulin during aging, but oxidative stress, calcium and b-amyloid (97, 98).
this effect seems well-evident only in men (84). Thus, the age-related decline of DHEAS might play a
In addition to the doubts concerning the patho- role in the occurrence of AD and other neurodegen-
physiology of the age-related decline of DHEAS secre- erative diseases.
tion, many questions still remain about the central and Beside the possible role of DHEA on cognitive
peripheral effects of adrenal androgens, since no functions, many studies suggest an involvement of
specific cellular or molecular targets for DHEA and the adrenal androgens also in the maintenance of
DHEAS have until now been identified (79). However, functional abilities (99, 100) and in the pathogenesis of
the results of DHEA administration in experimental several age-related diseases (101). Furthermore, the
animals or during clinical trials, suggest a pleiotropic reported lack of relationship between the age and the
effect of this hormone. DHEAS levels in subjects over 90 years old (102),
Indeed, DHEAS, but not DHEA, is able to activate the suggests that high DHEAS levels might be associated
hepatic peroxisome proliferator-activated receptor with a longer survival. Indeed, when age is factored out
alpha (85), an intracellular receptor of the steroid by appropriate polynomial regression, a significant
receptors family; this activation could modulate the component of DHEAS variations seems to exist (103).
fatty acid metabolism and the expression of peroxi- The data in the literature concerning DHEAS
somal enzymes, and thus could explain the DHEAS secretion in senile dementia are still conflicting (104±
anticarcinogenetic and chemoprotective effects (85). 110), although at the moment strong evidence points
Antidiabetic and antiobesity activities of DHEAS have to the possible role of DHEAS in limiting the cortisol
been also described in experimental animals, but they neurotoxic effect on hippocampal cells, in this way
are not always confirmed in humans. Furthermore, preventing the brain degeneration in AD (108).
high DHEAS levels have been related to a significant In a community-based study, no significant relation-
decrease of mortality for cardiovascular disease in men ships between DHEAS levels and cognitive function
but not in women (86). have been found (111). In another longitudinal study,
The most important findings, however, concern the lower DHEAS values were recorded in women with
effects of DHEAS on the CNS, where these steroids functional limitations, depressive symptomatology and
may be synthesized de novo, namely independently poor well-being, but not in men (112).
from the gonads and the adrenal glands, and thus Our results, obtained by the simultaneous evaluation
they are called `neurosteroids' (87). At the present it of the circadian rhythm of blood cortisol and DHEAS in
is unknown if changes in plasma DHEAS levels can physiological and pathological brain aging, including
directly affect CNS functions. However, at the CNS both AD and VD, indicated a clear decline in DHEAS
level, DHEAS and pregnenolone sulfate on the one secretion throughout the 24 h cycle in elderly subjects
hand and tetrahydroprogesterone, tetrahydrodeoxy- …n ˆ 23; age 76±96 years), and especially in old
corticosterone and androsterone on the other, effect demented patients …n ˆ 23; age 68±91 years), without

www.eje.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144 Aging and adrenal steroids 325

sex-related differences, in comparison with young


controls …n ˆ 10; age 25±32 years). Concerning the
type of dementia, patients with VD exhibited the lowest
DHEAS levels (Fig. 1).
The statistical analysis of our data by the population
mean cosinor method demonstrated the statistical
significance of the serum DHEAS circadian rhythm in
young and elderly subjects, even if with a marked
reduction of both mesor and amplitude in old subjects,
particularly if demented. However, a good maintenance
of the temporal relationship between the DHEAS and
cortisol crest-times persisted in both physiological and
pathological brain aging, as already described in
children and in young adults (113).
The molar ratio between cortisol and DHEAS levels
throughout the 24 h cycle was significantly higher in
elderly subjects, particularly if demented, in compar-
ison with young controls. Furthermore, old demented
patients exhibited a further increase of this ratio at
night-time, when compared with old controls, suggest-
ing the existence of a deeper neurotoxic effect of the
adrenocortical secretory imbalance (Fig. 1). Figure 2 Mean hippocampal volumes in physiological and
pathological brain aging and in young controls. Right: volume vs
This finding seems particularly interesting if we age, r ˆ 20:88; P , 0:001; vs cortisol nocturnal increase r ˆ 0:40;
consider that, in spite of the decline of DHEAS P , 0:05; vs DHEAS mesor, r ˆ 0:63; P , 0:01: Left: volume vs
secretion, well evident also in extreme senility, we age, r ˆ 20:82; P , 0:01; vs cortisol nocturnal increase, r ˆ 0:45;
have recently found similar cortisol levels in healthy P , 0:05; vs DHEAS mesor, r ˆ 0:64; P , 0:01: ***P , 0:001:
centenarians …n ˆ 20; age 100±106 years) and in
younger controls. Thus the cortisol/DHEAS molar ratio
was similar in centenarians and in healthy old controls hippocampal volumes and a progressive enlargement of
(data not shown). the ventricular size, which represents an index of
Due to the opposite changes of cortisol and androgen cerebral atrophy and volume loss (120). An important
secretions occurring in physiological and even more in finding to be emphasized is that the reduction of the
pathological brain aging, it seemed interesting to study, hippocampal volumes seems to be significantly related
by morphometric measures, the cerebral changes to the impairment of the cortisol nocturnal increase
occurring in these conditions, in order to try to and to the mean circadian value of serum DHEAS
correlate the neuroendocrinological features with the (Fig. 2). Even if in humans it is hard to demonstrate a
cerebral volumetric modifications. relationship between the hippocampal modifications
In particular we have considered the hippocampal and the adrenal secretion, our data suggest the
changes, since it is well known that both the age- existence of a link between the reduction of hippo-
related neuronal impairment and the degenerative campal volumes occurring with age and the trend to
changes of AD are especially evident in this area. increase of cortisol in the evening and at night-time
Indeed, the hippocampus is a particularly vulnerable and the decline of DHEAS secretion.
brain area, with a high expression of glucocorticoid Among the other cerebral volumes evaluated, only
receptors, and it is deeply involved in the regulation of the temporal lobe was significantly lower in AD
the HPA axis (114). Thus, it represents a vulnerable patients than in age-matched controls, suggesting the
link in the regulation of both HPA function and specificity for AD of these changes (121).
cognition (115).
Both in experimental animals and in humans, the
hippocampal volume decreases with aging, due not Conclusions
only to neuronal loss, but also to the effects of According to the results of our research and of many
neurotransmitters imbalance, excitatory amino acids studies in the literature, a clear dissociation of the
(116) and adrenal steroids (117, 118). To assess in vivo adrenocortical secretory pattern occurs with aging, due
the main cerebral volumes in healthy young and old to good maintenance of cortisol secretion and clear
subjects and in AD patients, we have recently impairment of that of androgens in elderly subjects,
performed a cerebral morphometric analysis by the with particular evidence in the demented ones.
magnetic resonance imaging method (119). Consequently, a significant increase of the cortisol/
In spite of the great interindividual variability, DHEAS molar ratio occurs in both physiological and
our study demonstrated an age-related decline in pathological aging.

www.eje.org
326 E Ferrari and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144

It is now well known that both glucocorticoids and acute adrenocorticotropin stimulation. Journal of Clinical Endo-
androgens have relevant effects on the brain, most crinology and Metabolism 1982 54 187±191.
9 Boscaro M, Paoletta A, Scarpa E, Barzon L, Fusaro P, Fallo F et al.
notably on the hippocampus, a critical area for learning Age-related changes in glucocorticoid fast feedback inhibition of
and memory, as well as a crucial center for the adrenocorticotropin in man. Journal of Clinical Endocrinology and
glucocorticoid inhibition of their own release (115, Metabolism 1998 83 1380±1383.
122, 123). The effects of the two kinds of steroids on the 10 Conforti N & Feldman S. Effects of dorsal fornix section and
hippocampectomy on adrenocortical responses to sensory
hippocampus are opposite, since cortisol promotes the stimulation in the rat. Neuroendocrinology 1976 22 1±7.
neuronal degeneration and death, whereas DHEAS 11 Sapolsky RM, Krey LC & McEwen BS. The neuroendocrinology
plays a protective role towards the structural damages of stress and ageing. The glucocorticoid cascade hypothesis.
and the functional impairment due to aging itself or to Endocrine Reviews 1986 7 284±301.
pathological situations, and stimulates the neuronal 12 Seeman TE & Robbins RJ. Aging and hypothalamic±pituitary±
adrenal response to challenge in humans. Endocrine Reviews
long-term potentiation (124). Thus, the age-related 1994 15 233±260.
decrease of DHEAS secretion necessarily results in an 13 McCarty R. Sympathetic-adrenal medullary and cardiovascular
impairment of the DHEAS-mediated antiglucocorticoid responses to acute cold stress in adult and aged rats. Journal of
activity, and consequently in an enhanced neurotoxicity the Autonomic Nervous System 1985 12 15±22.
14 Sapolsky RM. Stress, the Aging Brain and the Mechanisms of
of glucocorticoids and of excitatory amino acids (123). Neuron Death. Cambridge, MA: MIT Press, 1992.
As a whole our data suggest that, as in experimental 15 McEwen BS. Re-examination of the glucocorticoid hypothesis of
animals (125, 126), the brain aging may depend upon stress and aging. In Progress in Brain Research, Vol. 93: The
the cumulative exposure to increasing cortisol levels Human Hypothalamus in Health and Disease, pp 365±383. Eds DF
throughout life (32, 127), particularly if parallel to a Swaab, MA Hofman, M Mirmiran, R Ravid & FW van Leeuwen.
Amsterdam: Elsevier Science, 1992.
reduced androgen secretion. 16 Wilkinson CW, Peskind ER & Raskind MA. Decreased hypo-
The age-related hormonal changes, such as the thalamic±pituitary±adrenal axis sensitivity to cortisol feed-
opposite modifications of cortisol and melatonin secre- back inhibition in human aging. Neuroendocrinology 1997 65
tion, may play a role in the pathophysiology of the CNS 79±90.
17 Van Cauter E, Leproult R & Kupfer DJ. Effects of gender and age
degenerative changes also through the enhancement of on the levels and circadian rhythmicity of plasma cortisol.
5-lipoxygenase (5-LOX) expression in the CNS (128); Journal of Clinical Endocrinology and Metabolism 1996 81 2468±
this is the key enzyme responsible for the synthesis of 2473.
inflammatory leukotrienes, and probably involved in 18 Kern W, Dodt C, Born J & Fehm HL. Changes in cortisol and
neurodegeneration. Indeed, it is well known that 5-LOX growth hormone secretion during nocturnal sleep in the course
of aging. Journal of Gerontology Series A. Biological Sciences and
expression, prominent in hippocampal and cerebellar Medical Sciences 1996 51 M3±M9.
neurons, is stimulated by glucocorticoids (129) and 19 Hatzinger M, Z'Brunn A, Hemmeter U, Seifritz E, Baumann F,
depressed by melatonin (130). Therefore, the subtle but Holsboer-Trachsler E et al. Hypothalamic±pituitary±adrenal
significant changes of the hormonal balance occurring system function in patients with Alzheimer's disease. Neurobiol-
ogy of Aging 1995 16 205±209.
in aging may contribute to the onset and progression of 20 McEwen BE, Weiss JM & Schwartz LS. Selective retention of
the aging-associated neurodegenerative diseases. corticosterone by limbic structures in rat brain. Nature 1968
220 911±912.
21 Sapolsky RM & McEwen BS. Stress, glucocorticoids and their
role in degenerative changes in the aging hippocampus. In
References Treatment Development Strategies for Alzheimer's Disease, pp
1 McEwen BS. Protective and damaging effects of stress mediators. 151±172. Eds T Crook, R Bartus, S Ferris & C Flickers. New
New England Journal of Medicine 1998 338 171±179. Cannan, CT: Mark Powley Associates, 1986.
2 McEwen BS & Stellar E. Stress and the individual: mechanisms 22 Meaney M, Aitken D, Bhatnagar S, Van Berkel C & Sapolsky R.
leading to disease. Archives of Internal Medicine 1993 153 Postnatal handling attenuates neuroendocrine, anatomical and
2093±2101. cognitive impairments related to the aged hippocampus. Science
3 Swaab DF, Fliers E & Partiman TS. The suprachiasmatic nucleus 1988 238 766±768.
of the human brain in relation to sex, age and senile dementia. 23 Sapolsky R, Uno H, Rebert CS & Finch CE. Hippocampal damage
Brain Research 1985 342 37±44. associated with prolonged glucocorticoid exposure in primates.
4 Ball MJ. Neuronal loss, neurofibrillary tangles and granulova- Journal of Neuroscience 1990 10 2897±2902.
cuolar degeneration in the hippocampus with ageing and 24 Lupien S, Lecours AR, Lussoer I, Schwartz G, Nair NP &
dementia. Acta Neuropathologica 1977 37 111±118. Meaney M. Basal cortisol levels and cognitive deficits in human
5 Mani R, Lohr J & Jeste D. Hippocampal pyramidal cells and aging. Journal of Neuroscience 1994 14 2893±2903.
aging in the human: a quantitative study of neuronal loss in 25 Balladin J, Gottfries CG, Karlsson I, Lindstet G, Langstrom G &
sectors CA1 to CA4. Experimental Neurology 1986 94 29±40. Walinder J. Dexamethasone suppression test and serum
6 Moberg GP, Scapagnini U, de Groot J & Ganong WF. Effect of prolactin in dementia disorders. British Journal of Psychiatry
sectioning the fornix on diurnal fluctuation in plasma 1983 143 277±281.
corticosterone levels in the rat. Neuroendocrinology 1971 7 26 Lawlor BA, Tsuboyama G, Ryan TM, Mohs RC, Davis BM,
11±15. Davidson M et al. Agitation and postdexamethasone cortisol
7 Feldman S & Conforti N. Participation of the dorsal hippocam- levels in Alzheimer's disease. American Journal of Psychiatry
pus in the glucocorticoid feed-back effect on adrenocortical 1992 149 546±548.
activity. Neuroendocrinology 1980 30 42±55. 27 Davis KL, Davis BM, Greenwald BS, Mohs RC, MatheÁ AA,
8 Vermeulen A, Deslypere JP, Schelfhout W, Verdonck L & Johns CA et al. Cortisol and Alzheimer's disease: I. Basal studies.
Rubens R. Adrenocortical function in old age: response to American Journal of Psychiatry 1986 143 300±305.

www.eje.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144 Aging and adrenal steroids 327

28 Maeda K, Tanimoto K, Terada T, Shintani T & Kakigi T. Elevated disorders. Annals of the New York Academy of Sciences 1994 746
urinary free cortisol in patients with dementia. Neurobiology of 354±359.
Aging 1991 12 161±163. 48 Nichols NR. Glucocorticoids and aging. In Functional Endocri-
29 De Leon MJ, McRae T, Tsai JR, George AJ, Marcus DL, nology of Aging, vol 29, pp 1±26. Eds CV Mobbs & PR Hof. Basel:
Freedman M et al. Abnormal cortisol response in Alzheimer's Karger, 1998.
disease linked to hippocampal atrophy. Lancet 1988 ii 391±392. 49 Sadow TF & Rubin RT. Effects of hypothalamic peptides on the
30 Weiner MF, Vobach S, Olsson K, Svetlik D & Risser RC. Cortisol aging brain. Psychoneuroendocrinology 1992 17 293±314.
secretion and Alzheimer's disease progression. Biological 50 Holboer F, Spengler D & Heuser I. The role of corticotropin-
Psychiatry 1997 42 1030±1038. releasing hormone in the pathogenesis of Cushing's disease,
31 Meaney MJ, O'Donnel D, Rowe W, Tannenbaum B, Steverman A, anorexia nervosa, alcoholism, affective disorders and dementia.
Walker M et al. Individual differences in hypothalamic± Progress in Brain Research 1992 93 385±417.
pituitary±adrenal activity in later life and hippocampal aging. 51 Rasmuson S, Nasman B, Eriksson S, Carlstrom K & Olsson T.
Experimental Gerontology 1995 30 229±251. Adrenal responsivity in normal aging and mild to moderate
32 Lupien S, Lecours AR, Schwartz G, Sharma S, Hauger RL, Alzheimer's disease. Biological Psychiatry 1998 43 401±407.
Meaney MJ et al. Longitudinal study of basal cortisol levels in 52 Ferrari E, Borri R, Pontiggia B, Camozzi D, Spina ML, Stabile M
healthy elderly subjects: evidence for subgroups. Neurobiology of et al. Changes of the adrenocortical secretory pattern in
Aging 1996 17 95±105. physiological and pathological brain aging. Gerontologia 1998
33 Krieger DT, Silverberg AI, Rizzo F & Krieger HP. Abolition of 36 Suppl 1.
circadian periodicity of plasma 17-OH-cs levels in the cat. 53 Gottfries CG, Balldin J, Blennow K, Brane G, Karsson I, Regland B
American Journal of Physiology 1968 215 959±967. & Wallin A. Regulation of the hypothalamic±pituitary±adrenal
34 Ferrari E, Bossolo PA, Vailati A, Martinelli I, Rea A & Nosari I. axis in dementia disorders. Annals of the New York Academy of
Variations circadiennes des effects d'une substance vagolytique Sciences 1994 746 336±344.
sur le systeÁme ACTH-secreÂtant chez l'homme. Annales d'Endo- 54 Gottfries CG, Balldin J, Blennow K, Brane G, Karlsson I,
crinologie 1977 38 203±213. Regland B et al. Hypothalamic dysfunction in dementia. Journal
35 Dickson KL & Hasty DL. Effects of the pineal gland in unilaterally of Neural Transmitters 43 (Suppl) 1994 203±209.
adrenalectomized rats. Acta Endocrinologica 1972 70 438±442. 55 Keller-Wood ME & Dallman MF. Corticosteroid inhibition of
36 Wetteberg L. The relationship between the pineal gland and ACTH secretion. Endocrine Review 1984 5 1±24.
pituitary±adrenal axis in health, endocrine and psychiatric 56 Aneko M & Hiroshige T. Fast, rate sensitive corticosteroid
conditions. Psychoneuroendocrinology 1983 8 75±80. negative feed-back during stress. American Journal of Physiology
37 Sanchez De La Pena S, Halberg F & Hungar F. Pineal 1978 234 39±46.
chronomodulation. The feed-sideward. Clinical Chemistry News- 57 Armanini D. Corticosteroid receptors in lymphocytes: a possible
letter 1982 2 129±135. marker of brain involution? Journal of Steroid Biochemistry and
38 Milin J, Djakovic-Svaicer K & Demajo M. Rat pineal gland Molecular Biology 1994 49 429±434.
suppresses the injection stress-reactive ACTH outflow. Hormone 58 De Kloet ER. Brain corticosteroid receptor balance and
and Metabolic Research 1993 25 149±151. homeostatic control. Frontiers in Neuroendocrinology 1991 12
39 Magri F, Locatelli M, Balza G, Molla G, Cuzzoni G, Fioravanti M 95±164.
et al. Changes in endocrine circadian rhythms as markers of 59 Miller AH, Spencer RL, Pulera M, Kang S, McEwen BS & Stein M.
physiological and pathological brain aging. Chronobiology Adrenal steroid receptor activation in rat brain and pituitary
International 1997 14 385±396. following dexamethasone: implications for the dexamethasone
40 Ferrari E, Magri F, Locatelli M, Balza G, Nescis T, Battegazzore C suppression test. Biological Psychiatry 1992 32 850±869.
et al. Chrononeuroendocrine markers of the aging brain. Aging 60 De Kloet ER, Wallach G & McEwen BS. Differences in
1996 8 320±327. corticosterone and dexamethasone binding to rat brain and
41 Meneilly GS, Minaker KL, Young JB, Landesberg L & Rowe JW. pituitary. Endocrinology 1975 96 598±609.
Counterregulatory responses to insulin-induced glucose reduc- 61 Kovacs KJ & Makara GB. Corticosterone and dexamethasone act
tion in the elderly. Journal of Clinical Endocrinology and at different brain sites to inhibit adrenalectomy-induced
Metabolism 1985 61 178±182. adrenocorticotropin hypersecretion. Brain Research 1988 474
42 Raskind MA, Peskind ER & Wilkinson CW. Hypothalamic± 205±210.
pituitary±adrenal axis regulation and human aging. Annals of 62 Morano MI, Vazquez DM & Akil H. The role of hippocampal
the New York Academy of Sciences 1994 746 327±335. mineralocorticoid and glucocorticoid receptors in the hypotha-
43 Huizenga NA, Koper JW, De Lange P, Pols HA, Stolk RP, lamo±pituitary±adrenal axis of the aged Fisher rat. Molecular
Grobbee DE et al. Interperson variability but intraperson stability and Cellular Neuroscience 1994 5 400±412.
of baseline plasma cortisol concentrations, and its relation to 63 Guazzo EP, Kirkpatrick PJ, Goodyer IM, Shiers HM & Herbert J.
feedback sensitivity of the hypothalamo±pituitary±adrenal axis Cortisol, dehydroepiandrosterone (DHEA), and DHEA sulfate in
to a low dose of dexamethasone in elderly individuals. Journal of the cerebrospinal fluid of man: relation to blood levels and the
Clinical Endocrinology and Metabolism 1998 83 47±54. effects of age. Journal of Clinical Endocrinology and Metabolism
44 Heuser IJ, Gotthardt U, Schweiger U, Schmider J, Lammers C-H, 1996 81 3951±3960.
Dettling M et al. Age-associated changes of pituitary-adrenocor- 64 Walker MD, Henkin RI, Harlan AB & Caspar ATG. Distribution
tical hormone regulation in men: importance of gender. of tritiated cortisol in blood brain CSF and other tissues of the
Neurobiology of Aging 1994 15 227±231. cat. Endocrinology 1971 88 224±232.
45 Ferrari E, Solerte SB, Magri F, Fioravanti M, Magni P, Dori D et al. 65 Belanger A, Candas B, Dupont A, Cusan L, Diamond P &
Age-related changes of pituitary±adrenal axis in humans. In Gomez JL. Changes in serum concentrations of conjugated
Stress and the Aging Brain, pp 39±51. Ed. G Nappi. New York: and unconjugated steroids in 40 to 80-year old men.
Raven Press, 1990. Journal of Clinical Endocrinology and Metabolism 1994 79
46 Pavlov EP, Harman SM, Chrousos GP, Loriaux DL & 1086±1090.
Blackman MR. Responses of plasma adrenocorticotropin, 66 Carrol BJ, Feinberg M & Greden JF. A specific laboratory test for
cortisol, dehydroepiandrosterone to ovine corticotropin releas- diagnosis of melancholia: standardization, validation and
ing hormone in healthy aging men. Journal of Clinical clinical utility. Archives of General Psychiatry 1981 38 15±22.
Endocrinology and Metabolism 1986 62 767±772. 67 Ferrari E, Magri F, Dori D, Migliorati G, Nescis T, Molla G et al.
47 Holboer F, Grasser A, Friess E & Wiedemann K. Steroid effects on Neuroendocrine correlates of the aging brain in humans.
central neurons and implications for psychiatric and neurological Neuroendocrinology 1995 61 464±470.

www.eje.org
328 E Ferrari and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144

68 Sapolsky RM, Krey LC & McEwen BS. Prolonged glucocorticoid postmenopausal women [letter]. New England Journal of Medicine
exposure reduces hippocampal neuron number: implications for 1987 317±711.
aging. Journal of Neuroscience 1985 5 1221±1226. 87 Baulieu EE, Robel P, Vatier O, Haug A, Le Goascogne C &
69 Hornsby PJ. DHEA: a biologist's perspective. Journal of the Bourreau E. Neurosteroids: pregnenolone and dehydroepian-
American Geriatrics Society 1997 45 1395±1401. drosterone in the rat brain. In Receptor±Receptor Interaction. A
70 Guillemette C, Hum DW & Belanger A. Levels of plasma C19 New Intramembrane Integrative Mechanism, pp 89±104. Eds K
steroids and 5-alpha-reduced C19 steroid glucuridones in Fuxe & LF Agnati. Basingstoke: MacMillan, 1987.
primates, rodents, and domestic animals. American Journal of 88 Majewska MD. Neuronal actions of dehydroepiandrosterone.
Physiology 1996 271 E348±-E353. Possible roles in the brain development, aging, memory, and
71 Endoh A, Kristiansen SB, Casson PR, Busters JE & Hornsby PJ. affect. Annals of the New York Academy of Sciences 1995 774
The zona reticularis is the site of biosynthesis of dehydroepian- 111±120.
drosterone and dehydroepiandrosterone sulfate in the adult 89 Abadie J, Wright B, Correa G, Browne ES, Porter JR & Swec F.
human adrenal cortex, resulting from its low expression of Effects of dehydroepiandrosterone on neurotransmitter levels
3beta-hydroxysteroid dehydrogenase. Journal of Clinical Endocri- and appetite regulation of the obese Zucker rat. Diabetes 1993
nology and Metabolism 1996 81 3558±3565. 42 662±669.
72 Labrie F, Belanger A, Cusan L, Gomez JL & Candas B. Major decline 90 Roberts E, Bologa L, Flood JF & Smith GE. Effects of
in serum concentrations of adrenal C19 sex steroid precursors dehydroepiandrosterone and its sulfate on brain tissues in
and conjugated androgen metabolites during aging. Journal of culture and on memory in mice. Brain Research 1987 406 357±
Clinical Endocrinology and Metabolism 1997 82 2396±2402. 362.
73 Ohashi M, Kato K, Nawata H & Ibayashi H. Adrenocortical 91 Thijnssen JHH & Nieuwenhuyse H. DHEA: a Comprehensive
responsiveness to graded ACTH infusion in normal young and Review. New York: The Parthenon Publishing Group, 1999.
elderly human subjects. Gerontology 1986 32 43±51. 92 Monnet FP, Mahe V, Robel P & Baulieu EE. Neurosteroids, via
74 Kroboth PD, Salek FS, Pittenger AL, Fabian TJ & Frye RF. DHEA sigma receptors, modulate the [3H]norepinephrine release
and DHEAS: a review. Journal of Clinical Pharmacology 1999 39 evoked by N-methyl-d-aspartate in the rat hippocampus. PNAS
327±348. 1995 92 3774±3778.
75 Thomas G, Frenoy N, Legrain S, Sebag-Lanoe R, Baulieu EE & 93 Wolkowitz OM, Reus VI, Roberts E, Manfredi F, Chan T,
Debuire B. Serum dehydroepiandrosterone sulfate levels as an Ormitson S et al. Antidepressant and cognition-enhancing
individual marker. Journal of Clinical Endocrinology and Metabo- effects of DHEA in major depression. Annals of the New York
lism 1994 79 1273±1276. Academy of Sciences 1995 774 337±339.
76 Parker CR, Mixon RL, Brissie RM & Grizzle WE. Aging alters 94 Stanton PK & Survey JM. Depletion of norepinephrine, but not
zonation in the adrenal cortex of men. Journal of Clinical serotonin, reduces long-term potentiation in dentae gyrus of rat
Endocrinology and Metabolism 1997 82 3898±3901. hippocampal slices. Journal of Neuroscience 1986 5 2169±2176.
77 Hornsby PJ. Biosynthesis of DHEAS by the human adrenal 95 Sara SJ, Vankov A & Herve A. Locus coeruleus-evoked responses
cortex and its age-related decline. Annals of the New York in behaving rats. A clue to the role of noradrenaline in memory.
Academy of Sciences 1995 774 29±46. Brain Research Bulletin 1994 35 457±465.
78 Baulieu EE. Dehydroepiandrosteronea (DHEA): fountain of 96 Mao X & Barger SW. Neuroprotection by dehydroepiandroster-
youth? Journal of Clinical Endocrinology and Metabolism 1996 one-sulfate: role of an NFkB-like factor. NeuroReport 1998 9
81 3147±3151. 759±763.
79 Robel P & Baulieu EE. Dehydroepiandrosterone (DHEA) is a 97 Barger SW & Mattson MP. Induction of neuroprotective kappa B-
neuractive neurosteroid. Annals of the New York Academy of dependent transcription by secreted forms of the Alzheimer's beta
Sciences 1995 774 82±110. amyloid precursor. Molecular Brain Research 1996 40 116±126.
80 Clarke D, Fearon U, Cunnigham SK & McKenna TJ. The 98 Barger SW. Tumor necrosis factor: the good, the bad and the
steroidogenic effects of beta-endorphin and joining peptide: a umbra. In Neuroprotective Signal Transduction, pp 163±183. Ed.
potential role in the modulation of adrenal androgen produc- MP Mattson. Totawa, NJ, USA: Humana Press, 1997.
tion. Journal of Endocrinology 1996 151 301±307. 99 Rudman D, Shetty KR & Mattson DE. Plasma dehydroepian-
81 Parker LN, Lifrak E, Gelfand R, Lee J, Shively T & Kaplan V. drosterone sulfate in nursing home man. Journal of the American
Isolation, purification synthesis and binding of human adrenal Geriatric Society 1990 38 421±427.
gland cortical androgen stimulating hormone. Endocrine Journal 100 Morales AJ, Nolan JJ, Nelson JC & Yen SSC. Effects of
1993 1 441±445. replacement dose of dehydroepiandrosterone in men and
82 Penhoat A, Sanchez P, Jaillard C, Langlois D, Begeot M & Saez JM. women of advancing age. Journal of Clinical Endocrinology and
Human pro-opiomelanocortin (79±96) a proposed cortical Metabolism 1994 78 1360±1367.
androgen-stimulating hormone, does not affect steroidogenesis 101 Schneider EC & Reed JD. Life extension. New England Journal of
in cultured human adult adrenal cells. Journal of Clinical Medicine 1985 312 1159±1168.
Endocrinology and Metabolism 1991 72 23±26. 102 Ravaglia G, Morini P, Forti P, Maioli F, Boschi F, Mernardi M et al.
83 Mellon SH, Shively JE & Miller WL. Human pro-opiomelano- Anthropometric characteristics of healthy Italian nonagenarians
cortin (79±96) a proposed cortical androgen-stimulating and centenarians. British Journal of Nutrition 1997 77 9±17.
hormone, does not affect steroidogenesis in cultured human 103 Rotter JI, Wong FL, Lifrak ET & Parker LN. A genetic component
foetal adrenal cells. Journal of Clinical Endocrinology and to the variation of dehydroepiandrosterone sulfate. Metabolism
Metabolism 1991 72 19±21. 1985 34 731±736.
84 Beer NA, Jakubowicz DJ, Beer RM & Nestler JE. Disparate effects 104 Legrain S, Berr C, Frenoy N, Gourlet V, Bebuire B & Baulieu EE.
of insulin reduction with diltiazem on serum dehydroepian- Dehydroepiandrosterone sulfate in a long-term care aged
drosterone sulfate levels in obese, hypertensive men and women. population. Gerontology 1995 41 343±351.
Journal of Clinical Endocrinology and Metabolism 1994 79 1079± 105 Nasman B, Olsson T, Viitanen M & Carlstrom K. A subtle
1081. disturbance in the feedback regulation of the hypothalamic±
85 Peters JM, Zhou YC, Ram PA, Lee SS, Gonzales FJ & Waxman DJ. pituitary±adrenal axis in the early phase of Alzheimer's disease.
Peroxisome proliferator-activated receptor alpha required for Psychoneuroendocrinology 1995 20 211±220.
gene induction by dehydroepiandrosterone-3beta-sulfate. 106 Sunderland T, Merril CR, Harrington MG, Lawlor BA,
Molecular Pharmacology 1996 50 67±74. Molchan SE, Martinez R et al. Reduced plasma dehydroepian-
86 Barrett-Connor E & Khawt KT. Absence of an inverse relation of drosterone concentrations in Alzheimer's disease. Lancet 1989 ii
dehydroepiandrosterone sulfate with cardiovascular mortality in 570.

www.eje.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2001) 144 Aging and adrenal steroids 329

107 Leblhuber F, Windhanger E, Reisecker F, Steinparz FX & Dienstl V. cerebral morphometric correlates in normal aging and senile
Dehydroepiandrosterone sulfate in Alzheimer's disease. Lancet dementia. Dementia and Geriatric Cognitive Disorders 2000 11
1990 i 449. 90±99.
108 Leblhuber F, Windhager F, Steinparz FX, Dientsl E, Reisecker F & 120 De Leon MJ, Golomb J, George AE, Convit A, Tarshish CM,
Rittmannsberger H. Dehydroepiandrosterone-sulfate (DHEA-S) McRae T et al. The radiologic prediction of Alzheimer's disease:
serum levels in normal aging and Alzheimer's disease (AD). Age the atrophic hippocampal formation. American Journal of
1991 14 116±118. Neuroradiology 1993 14 897±906.
109 Schneider LS, Hinsey M & Lyness S. Plasma dehydroepiandros- 121 Double KL, Halliday GM, Kril JJ, Harasty JA, Cullen K, Beooks WS
terone in Alzheimer's disease. Biological Psychology 1992 31 et al. Topography of brain atrophy during normal aging and
205±208. Alzheimer's disease. Neurobiology of Aging 1996 17 513±521.
110 Regelson W & Kalimi M. Dehydorepiandrosterone (DHEA) ± the 122 Scheff SW, Scott SA & DeKoski ST. Quantification of synaptic
multifunctional steroid. II Effects on CNS, cell proliferation, density in the septal nuclei of young and aged Fischer 344 rats.
metabolic and vascular, clinical and other effects. Mechanism of Neurobiology of Aging 1991 12 3±12.
action. Annals of the New York Academy of Sciences 1994 719 123 McEwen BS. Stress and the aging hippocampus. Frontiers in
564±575. Neuroendocrinology 1999 20 49±70.
111 Dobbie JW. Adrenocortical nodular hyperplasias in ageing 124 Yoo A, Harris J & Dubrovsky B. Dose±response study of
adrenal. Journal of Pathology 1969 99 1±18. dehydroepiandrosterone sulfate on dentate gyrus long-term
112 Parker CR. Adrenal function in aging. Current Opinion in potentiation. Experimental Neurology 1996 137 151±156.
Endocrinology and Diabetes 1999 3 210±215. 125 Landfield PW & Elridge JC. The glucocorticoid hypothesis of
113 Haus H, Nicolau G, Lakatua DJ, Sackett-Lundeen L & Petrescu E. brain aging and neurodegeneration: recent modifications. Acta
Circadian rhythm parameters of endocrine functions in elderly Endocrinologica 1991 125 54±64.
subjects during the seventh to the ninth decade of life. 126 Landfield PW & Eldrige JC. The glucocorticoid hypothesis of age-
Chronobiology 1989 16 331±352. related hippocampal neurodegeneration: role of dysregulated
114 McEwen BS, de Leon MJ & Meaney MJ. Corticosteroids, the aging intraneuronal calcium. Annals of the New York Academy of
brain and cognition. Trends in Endocrinology and Metabolism Sciences 1994 746 308±321.
1999 10 92±96. 127 Lupien SJ & McEwen BS. The acute effects of corticosteroids on
115 Miller AH, Sastry G, Speranza AJ, Lawlor BA, Mohs RC, Ryan TM cognition: integration of animal and human model studies.
et al. Lack of association between cortisol hypersecretion and Brain Research Review 1997 24 1±27.
nonsuppression on the DST in patients with Alzheimer's disease. 128 Qu T, Uz T & Manev H. Inflammatory 5-LOX mRNA and protein
American Journal of Psychiatry 1994 151 267±270. are increased in brain of aging rats. Neurobiology of Aging 2000
116 McEwen BS, Albeck D, Cameron H, Chao HM, Gould E & 21 647±652.
Hastings N. Stress and the brain: a paradoxical role for adrenal 129 Zhang W, Lei ZM & Rao ChV. Immortalized hippocampal cells
steroids. In Vitamins and Hormones, pp 371±402. Ed. GD contain functional luteinizing hormone/human chorionic
Litwack. New York: Academic Press, 1995. gonadotropin receptors. Life Science 1999 65 2083±2098.
117 Galea LA & McEwen BS. Sex and seasonal differences in the rate 130 Steinhilberg D, Brungs M, Wertz O, Wiesenberg I, Danielsson C,
of cell proliferation in the dentate gyrus of adult meadow voles. Kahlen JP et al. The nuclear receptors for melatonin repress 5-
Neuroscience 1999 83 955±964. lipoxygenase gene expression in human B lymphocytes. Journal
118 Gould E, McEwen BS, Tanapat P, Galea LAM & Fuchs E. of Biological Chemistry 1995 270 7037±7040.
Neurogenesis in the dentate gyrus of the adult tree shrew is
regulated by psychosocial stress and NMDA. Journal of
Neuroscience 1997 17 2492±2498.
119 Magri F, Terenzi F, Ricciardi T, Fioravanti M, Solerte SB, Stabile M Received 15 May 2000
et al. Association between changes in adrenal secretion and Accepted 18 December 2000

www.eje.org

You might also like