Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Journal of Pathology

J Pathol 2007; 211: 173–180


Published online in Wiley InterScience
(www.interscience.wiley.com) DOI: 10.1002/path.2110

Review Article

The endocrine system and ageing


HS Chahal and WM Drake*
Department of Endocrinology, St Bartholomew’s Hospital, London, UK

*Correspondence to: Abstract


WM Drake, Department of
Endocrinology, St Bartholomew’s Complex changes occur within the endocrine system of ageing individuals. This article
Hospital, West Smithfield, explores the changes that occur in the metabolism and production of various hormones
London EC1A 7BE, UK. and discusses the resulting clinical consequences. As individuals age there is a decline
E-mail: w.m.drake@qmul.ac.uk in the peripheral levels of oestrogen and testosterone, with an increase in luteinizing
hormone, follicle-stimulating hormone and sex hormone-binding globulin. Additionally there
No conflicts of interest were
declared.
is a decline in serum concentrations of growth hormone, insulin-like growth factor-I and
dehydroepiandrosterone and its sulphate-bound form. Even though there are complex
changes within the hypothalmo–pituitary–adrenal/thyroid axis, there is minimal change
in adrenal and thyroid function with ageing. The clinical significance of these deficiencies
with age are variable and include reduced protein synthesis, decrease in lean body mass and
bone mass, increased fat mass, insulin resistance, higher cardiovascular disease risk, increase
in vasomotor symptoms, fatigue, depression, anaemia, poor libido, erectile deficiency and a
decline in immune function. For each endocrine system, studies have been carried out in
an attempt to reverse the effects of ageing by altering the serum hormonal levels of older
individuals. However, the real benefits of hormonal treatment in older individuals are still
being evaluated.
Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John
Wiley & Sons, Ltd.
Keywords: endocrine; ageing; follicle-stimulating hormone; luteinizing hormone; oestro-
gen; testosterone; growth hormone; thyroid; adrenal; dehydroepiandrosterone

Introduction endocrine function and the ageing process and explores


the age-related changes in hormone metabolism and
production, with their clinical consequences. In eval-
In ageing individuals, endocrine changes result in a
uating the changes that occur in endocrine function, it
decline in endocrine function involving the respon-
is important to distinguish between the real effects of
siveness of tissues as well as reduced hormone secre-
ageing on endocrine mechanisms from any confound-
tion from peripheral glands. This is coupled with
ing factors due to the higher prevalence of age-related
modifications in the central mechanisms controlling
illness.
the temporal organization of hormone release, with a
dampening of circadian hormonal and non-hormonal
rhythms. All endocrine glands are subject to the effects Menopause
of ageing and many endocrine functions are so inter-
twined that reduced function in one gland adversely By the mid-sixth decade of life, all women experience
affects the remainder. With the ageing process there the menopause. Ovulation frequency decreases by
are associated alterations in body composition and the age of 40, and reproductive ovarian function
a decline in functional status. Compared to younger ceases in the vast majority of women within the
individuals, healthy older individuals have decreased next 15 years [1]. In most women ovarian follicles
muscle mass, increased fat mass and decreased function less well during this period, with serum
strength. With healthy ageing there are changes in oestradiol concentrations being lower and follicle-
endocrine systems, including oestrogen (menopause), stimulating hormone (FSH) concentrations higher than
testosterone (andropause), growth hormone/insulin- in younger women. Luteinizing hormone (LH) is
like growth factor-I axis (somatopause), hypothala- unchanged [2]. Eventually follicular activity ceases,
mic–pituitary–thyroid axis, hypothalamic–pituitary– oestrogen concentrations fall to postmenopausal values
cortisol axis and dehydroepiandrosterone and its sul- and LH and FSH levels rise above premenopausal
phate (adrenopause). concentrations [1]. However, smaller amounts of a
This review article attempts to delineate some weaker oestrogen, oestrone, are still synthesized from
aspects of the interplay between the regulation of androstenedione in the cortex of the adrenal gland

Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
www.pathsoc.org.uk
174 HS Chahal et al

and in the interstitial ovarian cells [3]. Small amounts the menopause and certain aspects of cognition appear
of this oestrone can be transformed into oestradiol. to be related to changes in oestrogen. Observational
The changes in these serum concentrations result in studies and clinical trials have examined the influence
a series of further changes, including an increased of oestrogen on cognitive function, particularly mem-
risk of cardiovascular events, rapid loss of skeletal ory, in postmenopausal women, but the results are far
mass, vasomotor instability, psychological symptoms from consistent [14]. The vaginal mucosa atrophies in
and atrophy of oestrogen responsive tissue. postmenopausal women, which may lead to bleeding,
The risk of cardiovascular disease in premenopausal as the tissue is easily injured. In addition, oestrogen
women is lower than in men, but during the post- deprivation may lead to dysuria, urinary frequency
menopausal period the risk increases and is equal and incontinence. These symptoms may respond to
to males of equivalent age and risk factor profile. systemic or local oestrogen replacement therapy [15].
Prior to this increase in risk, serum concentrations of Causes of a loss in libido in postmenopausal women
atherogenic lipids deteriorate. Low-density lipoprotein may be due to a fall in both oestrogen and testosterone
and total cholesterol increase, whereas high-density levels as ovarian function stops [16].
lipoprotein decreases. This decrease in cardioprotec- There has been considerable debate about the
tive HDL is thought to be one of the causes of risk : benefit ratio of hormone replacement therapy,
increased coronary heart disease, myocardial infarction most notably with the recent publication of the results
and stroke in postmenopausal women [4]. Hormone from the Women’s Health Initiative (WHI) study.
replacement alters biochemical markers favourably, The study consisted of two parallel randomized,
but does not improve cardiovascular disease outcome double-blind, placebo-controlled clinical trials of hor-
[5,6]. mone therapy to determine whether conjugated equine
At the time of the menopause there is rapid loss oestrogen alone (for women with prior hysterectomy)
of bone, due to oestrogen withdrawal. This takes or in combination with progestin would reduce car-
place within the background of age-related bone loss diovascular events in mostly healthy postmenopausal
that begins in the fourth decade of life. In the women. The combined oestrogen and progestin com-
perimenopausal period, women lose 5–15% of their ponent of the WHI was stopped early, as women taking
bone mass, with 80% of this loss being trabecular hormonal therapy were determined to have an exces-
bone, which is more metabolically active than cortical sive risk of breast cancer [5]. At the time the trial was
bone [7]. There is a modest rise in serum ionized stopped, risks of coronary heart disease, stroke and
calcium without any change in parathyroid hormone pulmonary embolism were significantly increased in
(PTH), indicating a possible change in PTH set- the oestrogen + progestin group. Risks of colorectal
point which is reversed by hormone action. There cancer and of hip fracture were decreased and mortal-
is also a fall in the oestrogen-dependent components ity risk was not significantly different. The unopposed
of intestinal calcium absorption and renal tubular oestrogen arm was continued and the results were
reabsorption of calcium. The associated high bone similar to the combined conjugated equine oestrogen
resorption with normal PTH also suggests an increased arm in terms of heart disease, stroke and thromboem-
sensitivity of bone to PTH. There is no change in bolic events. A striking but not statistically signifi-
serum 1,25-vitamin D [8]. cant decrease in the incidence of breast cancer was
During the immediate menopausal period, when the observed [17]. Further analysis from the WHI study
rate of bone loss is greatest, oestrogen replacement showed that oestrogen therapy alone or in combina-
maintains bone mass and reduces fracture risk [9]. tion with progestin treatment increased the risk of both
Drugs that have been demonstrated to maintain bone dementia and mild cognitive impairment [18]. Another
mass in postmenopausal women include bisphospho- multicentre, prospective, controlled trial of oestrogen
nates, which act by inhibiting bone resorption more + progestin in postmenopausal women with estab-
than formation [10], and raloxifene, which is a selec- lished coronary disease failed to support the use of
tive oestrogen receptor modulator acting selectively on hormone replacement therapy for the secondary pre-
bone and lipid profiles [11]. vention of heart disease [6]. Patients need to under-
Vasomotor symptoms originate in the hypothalamus, stand that recent studies have shown that hormone
with a resetting and narrowing of the thermoregula- replacement therapy can carry an increased risk of
tory system [1]. The hot flush is preceded by an LH ischaemic stroke, coronary events, venous thrombo-
surge, although there is no associated change in serum sis and possibly breast cancer. In order to minimize
oestradiol [4]. Decreased oestrogen levels may reduce these hazards, hormone replacement therapy should be
serotonin levels and thus cause an up-regulation of considered only for severe menopausal symptoms and
the 5-HT2A receptor in the hypothalamus. Additional for the shortest possible time in women who are fully
serotonin is released, which causes activation of the informed of these risks [19].
5-HT2A receptor, thus changing the set point tem-
perature and resulting in hot flushes [12]. However, Andropause
the full mechanism is not completely understood. Hor-
mone replacement reduces but does not eliminate such Ageing is associated with changes in gonadal steroid
episodes [13]. Cognitive disturbances are reported at production in men as well as women. As the

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The endocrine system and ageing 175

population is living to an older age, there has been there is still no consensus as to whether androgen
much interest in the study of the ageing male with treatment is beneficial to men over 50. Much of the
reference to so called ‘rejuvenating hormones’, in par- uncertainty is due to the brief duration of many of the
ticular androgens. For many years there was much protocols, such that the effects of prolonged testos-
debate as to whether serum total testosterone levels terone replacement are not clear.
were truly lower in healthy older men, or whether this
decline was attributable to the ageing process, with the
observed decline occurring as a result of confound- Growth hormone–insulin-like growth
ing effects due to chronic illness and medications. factor-I axis
However, from cross-sectional and longitudinal stud-
ies there is now agreement that in healthy men there Growth hormone (GH) is both anabolic and lipolytic
is a gradual but progressive age-dependent decline and the action of growth hormone on peripheral tissues
in testosterone levels, termed the andropause [20,21]. is mediated, in part, by circulating (hepatic-generated)
This is more marked for free testosterone than for total or paracrine insulin-like growth factor-I (IGF-I) [34].
testosterone, due to an age-associated increase of sex The secretion of GH undergoes dramatic changes
hormone-binding globulin levels [22]. The age-related during life. GH output is relatively low before puberty,
decline in testosterone level does not start at any spe- but with sexual maturation and adolescence there is
cific point in older subjects and it varies from modest a period of high GH output and accelerated somatic
to severe (with unclear clinical consequences), which growth [35]. With ageing, numerous studies have
is different from the sharp reduction of oestrogen pro- shown that GH secretion and serum GH concentrations
duction in females at the menopause [23]. fall, both basally and in response to stimuli, and this is
The decline in serum testosterone concentrations paralleled by a decline in IGF-I [34]. GH production
is mainly due to decreased production rates in older and IGF-I concentrations decline by more than 50% in
men [24] and this is a result of abnormalities at healthy older adults [36]. The progressive decline in
all levels of the hypothalamic–pituitary–testicular GH secretion has been termed the ‘somatopause’. In
axis [25]. In longitudinal studies, serum LH and older subjects the decrease in GH secretion is known
FSH levels show an age-related increase. However, to cause a reduction of protein synthesis, a decrease
serum LH concentrations often do not reciprocate the in lean body mass and bone mass and a decline in
decline in testosterone with age [26] — most likely immune function [34].
a result of impaired gonadotrophin-releasing hormone The neuroendocrine mechanisms of the somatopause
secretion and alterations in gonadal steroid feedback are uncertain. Early studies suggesting senescent
mechanisms [27]. The testosterone response to LH and changes in the pituitary [37,38] have not been sup-
human chorionic gonadotrophin decreases with ageing ported by the observations that there is no decrease
[28] and the circadian rhythm of plasma testosterone in the number of pituitary somatotroph cells [39] or
secretion, with higher levels in the morning than in that exogenous growth hormone-releasing hormone
the evening, is generally lost in older men [29]. (GHRH) [40,41] or GH-releasing peptide analogues
The clinical features associated with reduced testos- [42] are able to rejuvenate GH output and plasma IGF-
terone levels in ageing men include increased fat mass, I levels in older individuals. Consequently, attention
loss of muscle and bone mass, fatigue, depression, has shifted to potential alterations of the hypothalamic
anaemia, poor libido, erectile deficiency [20], insulin regulation of GH secretion, with data suggesting an
resistance [30] and higher cardiovascular risk [31]. age-dependent decrease in endogenous hypothalamic
These are similar to changes associated with testos- GHRH output, contributing to the age-associated GH
terone deficiency in young men, so the syndrome of decline [43]. Low physical fitness and higher adiposity
androgen deficiency of the ageing male (ADAM) has in older individuals also contributes to the decreased
been proposed. However, each of these clinical fea- GH secretion [44], although the mechanisms underly-
tures may also occur in older men with normal andro- ing these observations are not clear. Low IGF-I levels
gen levels and so the ADAM syndrome has not been reflect decreased GH secretion rather than a loss of
universally accepted [32]. hepatic responsiveness to the hormone, as circulating
As there is a certain proportion of middle-aged and IGF-I levels increase similarly in young and old men
older men with serum total testosterone levels below after exogenous administration of either GH or GHRH
the reference range for young adult males, there is [34].
a suggestion that supplementing testosterone in older In younger GH-deficient adults there are alterations
men with low testosterone levels into a range that is in body composition, including physical performance,
mid-normal for healthy, young men may prevent or psychological well-being and substrate metabolism,
reverse the effects of ageing [33]. Over the last decade, which resemble the ageing phenotype. These features
several clinical studies have been undertaken to deter- are improved by long-term hormone replacement with
mine whether testosterone supplementation in ageing recombinant human GH [45]. This had led to the
is beneficial. Despite several trials examining vari- suggestion that the elderly have genuine GH deficiency
ous parameters, including body composition, muscle and, by implication, would benefit from GH treatment.
strength, bone density, metabolism and lipid profile, There are many unanswered questions about the use

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
176 HS Chahal et al

of GH in older individuals. A study published in 1990 Hypothalmo–pituitary–thyroid axis


showed that 6 months treatment of recombinant GH
in 12 healthy 61–81 year-old men who had serum The hypothalmo–pituitary–thyroid axis undergoes a
IGF-I concentrations below those of healthy younger significant number of complex physiological alter-
men resulted in an increase in lean body mass by 9% ations associated with ageing. However, direct age-
and a decrease in adipose tissue mass by 15% [46]. related changes need to be distinguished from indirect
However, the weekly dose of GH was approximately alterations caused by simultaneous thyroid or non-
twice as high as the dose used in non-elderly GH- thyroidal illness, or other physiological or pathophys-
deficient adults, the study was not double-blinded iological states whose incidence increases with age.
and there was no assessment of muscle strength, Several changes formerly believed to be a direct result
exercise endurance or quality of life [47]. A double- of the ageing process have subsequently been shown to
blind placebo-controlled study showed that GH with be due to the increased prevalence of subclinical thy-
or without sex steroids in healthy women (n = 57) roid disease and/or the result of non-thyroidal illness.
and men (n = 74) aged 65–88 years increased lean This makes interpretation of thyroid function tests dif-
body mass and decreased fat mass [48]. However, ficult in the elderly [53]. Thyroid hormone clearance
there was no change in muscle strength or maximal decreases with age, but thyroid hormone secretion
oxygen uptake during exercise. These findings were is also reduced, leading to unchanged total and free
similar to a previous randomized, controlled, double- serum thyroxine (T4) concentrations [54]. In contrast
blind trial in 1996, in which 52 healthy men aged to thyroxine, serum total and free triiodothyronine (T3)
>69 years with well-preserved functional ability but concentrations decrease with ageing. This reduction is
low baseline IGF-1 levels were given 6 months of believed to be mostly due to reduced peripheral con-
physiological doses of GH [49]. Body composition version of T4 to T3, due either to the direct effect of
improved but functional ability did not. A further study non-thyroidal illness or to ageing itself [53].
recruited 18 healthy older men (aged 65–82 years) In older, apparently euthyroid patients, serum
who initially underwent progressive weight training thyroid-stimulating hormone (TSH) concentrations
for 14 weeks to invoke a trained state, then were may be reduced [55,56], but this is usually a patholog-
randomized to receive GH or placebo while continuing ical finding indicating either exogenous or endogenous
a further 10 weeks of strength training [50]. The thyrotoxicosis [57]. However, in elderly patients with-
out clinical or subclinical hyperthyroidism, slightly
results suggested that supplementation with GH does
decreased serum TSH is seen [56,58]. An age-
not augment the response to strength training in
dependent reduction of daily TSH secretion rate has
older men.
been reported [59]. The reason for such age-dependent
Data have also suggested that the age-related decline
reduction of TSH secretion is uncertain. It may be
in testosterone seen in men may contribute to the
due to supersensitivity of thyrotrophs to the neg-
reduction in GH secretion; thus, testosterone may act
ative feedback from T4, but other theories, such
synergistically with GH in reversing this GH secretion as reduced hypothalamic thyroid-releasing hormone
decline. Non-pharmacological doses of combined GH secretion, have not been excluded [53]. The ampli-
and testosterone in older men have been shown to tude of the nocturnal pulses of TSH secretion, which
improve selected aspects of physical performance results in the majority of 24 h TSH secretion, is lower
and increased muscle IGF-I gene expression without in older subjects [60]. This decrease probably results
measurably changing body composition or muscle in decreased T4 secretion in response to the decrease
strength [51]. In a more recent study, co-administration in T4 clearance in older individuals [54].
of low-dose GH with testosterone resulted in beneficial The prevalence of thyroid disease increases with
changes in mid-thigh muscle and aerobic capacity [52]. age and all forms of thyroid disease are encountered.
The initial enthusiasm for the potential benefits of However, the clinical manifestations are different from
GH replacement in aged individuals has been severely those encountered in younger patients. In the elderly,
dampened by its known adverse side-effects, including autoimmune hypothyroidism is particularly prevalent.
arthralgia, carpal tunnel syndrome, oedema and hyper- Hyperthyroidism is mainly characterized by cardiovas-
glycaemia. There are also particular concerns over the cular symptoms and is frequently due to toxic nodular
links between the GH–IGF–I axis and the develop- goitres. Thyroid carcinoma is also more aggressive
ment of cancer in the normal population [47,48]. Stud- [61]. Ageing is also associated with the appearance of
ies to date have been for a maximum of 12 months, thyroid autoantibodies, but the biological and clinical
so long-term safety data are not available. Currently significance of this is still unknown. Some data have
there is no ‘magic pill’ that reverses the process of shown that these thyroid autoantibodies are rare in
ageing, and GH therapy for ‘anti-ageing’ has currently healthy centenarians and in other highly selected aged
not been proved to be effective [47]. Long-term stud- populations, whereas they are frequently observed in
ies are required to determine the efficacy and safety of unselected or hospitalized elderly patients, thus sug-
GH treatment in older adults who are not GH-deficient. gesting that these autoantibodies are not the conse-
It remains to be seen whether GH secreatagogues are quence of the ageing process itself, but rather are
beneficial in the elderly. related to age-associated disease [62].

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The endocrine system and ageing 177

A major, unresolved issue is whether and to what individuals might also be regulated by the HPA axis
extent the complex physiological changes seen in [71]. Furthermore, in both men and women cortisol
the hypothalmo–pituitary–thyroid axis contributes to levels are strongly associated with a risk of clinical
the pathogenesis of age-associated diseases such as fractures [72]. Lastly, there is an association between
atherosclerosis, coronary heart disease and neurologi- 24 h cortisol production rate and increased body fat
cal disorders [53]. in older men. Thus, the increase in HPA axis activity
may play a role in the alterations in body composition
and central fat distribution that are seen in ageing [73].
Hypothalmo–pituitary–adrenal axis

The hypothalamo–pituitary–adrenal (HPA) axis is Dehydroepiandrosterone


involved in life-sustaining homeostatic and allostatic
adjustments to internal and external stressors. This Dehydroepiandrosterone (DHEA) and its sulphate-
stress-adaptive axis is a dynamic feedback network bound form (DHEAS) are the most abundant steroid
with circadian rhythmicity and pulsatile neurohormone hormones, although their physiological functions have
secretion [63]. However, how ageing causes changes not been fully delineated. As well as an abdundant cir-
in the axis is incompletely understood. With age there culating adrenal androgen, DHEA(S) is also thought
are variable changes in the effects of cortisol on ACTH to act directly as a neurosteroid that may have car-
secretion or of ACTH on cortisol secretion [64]. How- dioprotective, antidiabetic, anti-obesity and immuno-
ever, there appears to be no deficiency of adrenal enhancing properties [74]. There has been much debate
production of corticosteroids in ageing [65]. Healthy on the anti-ageing properties of DHEA and its poten-
ageing likely disrupts neuroendocrine mechanisms that tial as a ‘hormone of youth’ [75]. Unlike the rela-
coordinate within axis pulsatile and 24 h rhythmic cor- tively unaffected cortisol biosynthesis, the major age-
tisol release, and also alters the inter-axis mechanisms related change in the human adrenal cortex is a striking
that link LH and cortisol release. In older subjects, decrease in the biosynthesis of DHEA(S) [66,76,77].
serum cortisol secretion concentrations may vary more The blood level of DHEA, most of which is present in
within a 24 h period as compared to younger subjects the sulphated form (DHEAS), peaks at approximately
[63]. There is a 20–50% increase in 24 h mean cor- 20 years of age and declines rapidly and markedly
tisol levels between 20 and 80 years of age [66]. The after the age of 25 [76]. By the age of 80, patients
evening nadir in serum cortisol concentrations may be have DHEA levels 10–20% of those of younger coun-
higher and earlier in older subjects [66,67]. Levels of terparts [78]. Histomorphological analysis of adrenal
corticosteroid binding globulin have not been shown to specimens suggests that ageing results in alterations
alter with age [68]. With dexamethasone, inhibiton of within the adrenal cortex, resulting in a reduction in
ACTH and cortisol secretion is similar to younger indi- the size of the zona reticularis, this being responsible
viduals [64], but this inhibition may be slower in onset for the diminished production of DHEA [79].
[69]. In older women, serum cortisol concentrations The physiological consequences of a decline in
increase more with exogenous ACTH [64]. The rise DHEA with age are not fully understood. Many have
in serum concentrations in fasting older and younger speculated that administration of DHEA may reverse
men are similar and serum cortisol response to stress ageing effects and there is widespread commercial
is prolonged in older individuals [63]. Several studies availability of DHEA outside the regular pharmaceu-
have shown gender-specific, age-related alterations in tical networks, without adequate scientific evidence
the HPA axis. With healthy ageing there is an increase [80]. Cross-sectional studies have noted an association
in the cortisol response to challenge from CRH and between the decline in DHEAS levels and cardiovas-
diminished hypothalamic–pituitary sensitivity to glu- cular disease, breast cancer, low bone mineral density,
cocorticoid feedback inhibition. However, this is more depressed mood, type 2 diabetes and Alzheimer’s dis-
profound in older women than older men [68]. ease. However, this may reflect the ageing process per
Age-related changes in the HPA axis may have far- se rather than there being a causal relationship [81].
reaching physiological significance. There is growing This age-related decline in circulating DHEA(S)
evidence supporting the view that chronic cortisol has led to a number of randomized trials assessing
excess may lead to hippocampal atrophy and cognitive the effect of oral DHEA in otherwise healthy older
impairment during ageing. The alterations in cortisol subjects. In the first randomized placebo-controlled
circadian amplitude and phase could be involved in cross-over trial, 50 mg DHEA was administered to 13
the aetiology of sleep disorders in the elderly [66]. men and 17 women aged 40–70 years for 6 months.
Additionally, in older females increasing levels of There was an improvement in well-being and no
HPA axis activity, as measured by urinary free cortisol change in insulin sensitivity or body composition.
excretion, are associated with a decline in memory Bioavailable IGF-I increased slightly, whereas HDL
performance [70]. In healthy older men, cortisol levels cholesterol decreased in women [82]. In the largest
are inversely related to bone mineral density and study to date, a double-blind randomized parallel study
the rate of bone loss, suggesting that bone density of 140 men and 140 women aged 60–79 years, who
and the rate of involutional bone loss in healthy were given 50 mg DHEA or placebo daily, showed

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
178 HS Chahal et al

no improvement in well-being or cognition [83]. In aged-related changes is ultimately beneficial. So far


the study, women >70 years had increased libido, and research has not found the ‘magic pill’ to reverse the
slight but significant gains in bone mineral density process of ageing and the quest for a ‘hormone of
were observed in women but not in men. Other trials youth’ still carries on.
have failed to demonstrate any benefit of DHEA
on well-being, mood, cognition or activities of daily
living [84–86]. From these studies, it can be concluded References
that the decline in DHEA concentrations does not
1. Johnson SR. Menopause and hormone replacement therapy. Med
necessarily lead to impaired well-being, cognition or Clin North Am 1998;82(2):297–320.
sexuality per se [80]. 2. Sherman BM, West JH, Korenman SG. The menopausal transi-
In healthy ageing there is a marked sexual dimor- tion: analysis of LH, FSH, estradiol, and progesterone concentra-
phism in adrenal hormone regulation. In older women, tions during menstrual cycles of older women. J Clin Endocrinol
Metab 1976;42(4):629–636.
lower DHEA(S) and higher cortisol levels are seen
3. Longcope C, Jaffee W, Griffing G. Metabolic clearance rates
compared to older men, which persists into advanced of androgens and oestrogens in ageing women. Maturitas
age. This is in contrast to cortisol levels in men and 1980;2(4):283–290.
women, which show a progressive, parallel increase 4. Speroff L, Glass RH, Kase NG (eds). Clinical Gynecologic
with ageing. The consequence of sexual dimorphism Endocrinology and Infertility (6th edn). Lippincott Williams and
Wilkins: Baltimore, MD, 1999.
in adrenal hormones may have implications for age-
5. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooper-
related changes in cardiovascular disease, brain func- berg C, Stefanick ML, et al. Risks and benefits of estrogen plus
tion and bone metabolism [68]. progestin in healthy postmenopausal women: principal results from
In summary, there is no clear clinical consequence the Women’s Health Initiative randomized controlled trial. J Am
of the age-related decrease in serum concentration Med Assoc 2002;17(3):321–333.
6. Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B,
of DHEA and there are no clear benefits of DHEA et al. Randomized trial of estrogen plus progestin for secondary
replacement in older individuals [80]. prevention of coronary heart disease in postmenopausal women.
Heart and Estrogen/progestin Replacement Study (HERS)
Research Group. J Am Med Assoc 1998;280(7):605–613.
Conclusion 7. Riggs BL, Melton LJI. Involutional osteoporosis. N Engl J Med
1986;314(26):1676–1686.
8. Nordin BE, Wishart JM, Clifton PM, McArthur R, Scopacasa F,
Complex changes are seen in many endocrine sys- Need AG, et al. A longitudinal study of bone-related bio-
tems with ageing that occur independently of factors chemical changes at the menopause. Clin Endocrinol (Oxf)
associated with the higher prevalence of age-related 2004;61(1):123–130.
illnesses. Endocrine deficiencies in older individuals 9. Kiel DP, Felson DT, Anderson JJ, Wilson PW, Moskowitz MA.
Hip fracture and the use of estrogens in postmenopausal women.
include a decrease in the peripheral levels of oestro- The Framingham Study. N Engl J Med 1987;317(19):1169–1174.
gen and testosterone, with an increase in LH, FSH 10. Liberman UA, Weiss SR, Broll J, Minne HW, Quan H, Bell NH,
and sex hormone-binding globulin. In addition there et al. Effect of oral alendronate on bone mineral density and
is a decline in serum concentrations of GH, IGF-I and the incidence of fractures in postmenopausal osteoporosis. The
DHEA(S). The endocrine functions that are essential Alendronate Phase III Osteoporosis Treatment Study Group.
N Engl J Med 1995;333(22):1437–1443.
to life, such as adrenal and thyroid functions, show 11. Delmas PD, Bjarnason NH, Mitlak BH, Ravoux AC, Shah AS,
a minimal overall change in basal levels with ageing, Huster WJ, et al. Effects of raloxifene on bone mineral density,
even though there are complex changes that do occur serum cholesterol concentrations, and uterine endometrium in
within the hypothalmo–pituitary–adrenal/thyroid axis. postmenopausal women. N Engl J Med 1997;337(23):1641–1647.
The clinical significance of these deficiencies with 12. ACOG Task Force on Hormone Therapy. Obstetrics and
Gynecology 2004;104(S4):112S.
age are variable and are still being evaluated. The 13. Hammar M, Christau S, Nathorst-Boos J, Rud T, Garre K. A
menopause results in a series of changes in lipid double-blind, randomized trial comparing the effects of tibolone
metabolism, bone loss, vasomotor symptoms and pos- and continuous combined hormone replacement therapy in
sible changes in cognition. Likewise, declines in postmenopausal women with menopausal symptoms. Br J Obstet
gonadal function in men are associated with increased Gynaecol 1998;105(8):904–911.
14. Shaywitz SE, Shaywitz BA, Pugh KR, Fulbright RK, Skud-
fat mass, loss of muscle and bone mass, fatigue, larski P, Mencl WE, et al. Effect of estrogen on brain activation
depression, anaemia, poor libido, erectile deficiency, patterns in postmenopausal women during working memory tasks.
insulin resistance and higher cardiovascular disease J Am Med Assoc 1999;281(13):1197–1202.
risk. Similarly, a decline in the GH–IGF–I axis results 15. Hendrix SL, Cochrane BB, Nygaard IE, Handa VL, Barnabei VM,
in reduced protein synthesis, decrease in lean body Iglesia C, et al. Effects of estrogen with and without progestin on
urinary incontinence. J Am Med Assoc 2005;293(8):935–948.
mass and bone mass and a deterioration in immune 16. Shifren JL, Braunstein GD, Simon JA, Casson PR, Buster JE,
function. Changes in adrenal hormones have variable Redmond GP, et al. Transdermal testosterone treatment in women
clinical significance. with impaired sexual function after oophorectomy. N Engl J Med
For each endocrine system there have been many 2000;343(10):682–688.
studies trying to reverse the effects of ageing by 17. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA,
Black H, et al. Effects of conjugated equine estrogen in
restoring the serum hormonal levels of older indi- postmenopausal women with hysterectomy: the Women’s
viduals back into ‘younger ranges’. However, cur- Health Initiative randomized controlled trial. J Am Med Assoc
rently it is unclear whether treatment of many of these 2004;291(14):1701–1712.

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
The endocrine system and ageing 179

18. Shumaker SA, Legault C, Kuller L, Rapp SR, Thal L, Lane DS, 39. Dobado-Berrios PM, Ruiz-Navarro A, Almaden Y, Malagon MM,
et al. Conjugated equine estrogens and incidence of probable Garrido JC, Ramirez-Gutierrez JL, et al. Heterogeneity of growth
dementia and mild cognitive impairment in postmenopausal hormone (GH)-producing cells in aging male rats: ultrastructure
women: Women’s Health Initiative Memory Study. J Am Med and GH gene expression in somatotrope subpopulations. Mol Cell
Assoc 2004;291(24):2947–2958. Endocrinol 1996;118(1–2):181–191.
19. Crawford F, Langhorne P. Time to review all the evidence for 40. Corpas E, Harman SM, Pineyro MA, Roberson R, Blackman MR.
hormone replacement therapy. Br Med J 2005;330(7487):345. Continuous subcutaneous infusions of growth hormone (GH)
20. Gray A, Feldman HA, McKinlay JB, Longcope C. Age, disease, releasing hormone 1–44 for 14 days increase GH and insulin-
and changing sex hormone levels in middle-aged men: results of like growth factor-I levels in old men. J Clin Endocrinol Metab
the Massachusetts Male Aging Study. J Clin Endocrinol Metab 1993;76(1):134–138.
1991;73(5):1016–1025. 41. Corpas E, Harman SM, Pineyro MA, Roberson R, Blackman MR.
21. Morley JE, Kaiser FE, Perry HMI, Patrick P, Morley PM, Growth hormone (GH)-releasing hormone-(1–29) twice daily
Stauber PM, et al. Longitudinal changes in testosterone, luteinizing reverses the decreased GH and insulin-like growth factor-I levels
hormone, and follicle-stimulating hormone in healthy older men. in old men. J Clin Endocrinol Metab 1992;75(2):530–535.
Metabolism 1997;46(4):410–413. 42. Chapman IM, Hartman ML, Pezzoli SS, Thorner MO. Enhance-
22. Harman SM, Metter EJ, Tobin JD, Pearson J, Blackman MR. ment of pulsatile growth hormone secretion by continuous infusion
Longitudinal effects of aging on serum total and free testosterone of a growth hormone-releasing peptide mimetic, L-692,429, in
levels in healthy men. Baltimore Longitudinal Study of Aging. older adults — a clinical research center study. J Clin Endocrinol
J Clin Endocrinol Metab 2001;86(2):724–731. Metab 1996;81(8):2874–2880.
23. Vermeulen A. Androgen replacement therapy in the aging 43. Russell-Aulet M, Jaffe CA, Mott-Friberg R, Barkan AL. In vivo
male — a critical evaluation. J Clin Endocrinol Metab semiquantification of hypothalamic growth hormone-releasing hor-
2001;86(6):2380–2390. mone (GHRH) output in humans: evidence for relative GHRH defi-
24. Ishimaru T, Pages L, Horton R. Altered metabolism of androgens ciency in aging. J Clin Endocrinol Metab 1999;84(10):3490–3497.
in elderly men with benign prostatic hyperplasia. J Clin Endocrinol 44. Vahl N, Jorgensen JO, Jurik AG, Christiansen JS. Abdominal
Metab 1977;45(4):695–701. adiposity and physical fitness are major determinants of the age
25. Mulligan T, Iranmanesh A, Johnson ML, Straume M, Veld- associated decline in stimulated GH secretion in healthy adults.
huis JD. Aging alters feed-forward and feedback linkages between J Clin Endocrinol Metab 1996;81(6):2209–2215.
LH and testosterone in healthy men. Am J Physiol 1997;273(4, pt 45. Carroll PV, Christ ER, Bengtsson BA, Carlsson L, Christiansen
2):R1407–1413. JS, Clemmons D, et al. Growth hormone deficiency in adulthood
26. Mitchell R, Hollis S, Rothwell C, Robertson WR. Age related and the effects of growth hormone replacement: a review.
changes in the pituitary–testicular axis in normal men; lower Growth Hormone Research Society Scientific Committee. J Clin
serum testosterone results from decreased bioactive LH drive. Clin Endocrinol Metab 1998;83(2):382–395.
Endocrinol (Oxf) 1995;42(5):501–507. 46. Rudman D, Feller AG, Nagraj HS, Gergans GA, Lalitha PY,
27. Veldhuis JD, Zwart A, Mulligan T, Iranmanesh A. Muting of Goldberg AF, et al. Effects of human growth hormone in men
androgen negative feedback unveils impoverished gonadotropin- over 60 years old. N Engl J Med 1990;323(1):1–6.
releasing hormone/luteinizing hormone secretory reactivity in 47. Vance ML. Can growth hormone prevent aging? N Engl J Med
healthy older men. J Clin Endocrinol Metab 2001;86(2):529–535. 2003;348(9):779–780.
28. Harman SM, Tsitouras PD. Reproductive hormones in aging men. 48. Blackman MR, Sorkin JD, Munzer T, Bellantoni MF, Busby-
I. Measurement of sex steroids, basal luteinizing hormone, and Whitehead J, Stevens TE, et al. Growth hormone and sex steroid
Leydig cell response to human chorionic gonadotropin. J Clin administration in healthy aged women and men: a randomized
Endocrinol Metab 1980;51(1):35–40. controlled trial. J Am Med Assoc 2002;288(18):2282–2292.
29. Bremner WJ, Vitiello MV, Prinz PN. Loss of circadian rhythmic- 49. Papadakis MA, Grady D, Black D, Tierney MJ, Gooding GA,
ity in blood testosterone levels with aging in normal men. J Clin Schambelan M, et al. Growth hormone replacement in healthy
Endocrinol Metab 1983;56(6):1278–1281. older men improves body composition but not functional ability.
30. Abate N, Haffner SM, Garg A, Peshock RM, Grundy SM. Sex Ann Intern Med 1996;124(8):708–716.
steroid hormones, upper body obesity, and insulin resistance. J 50. Taaffe DR, Pruitt L, Reim J, Hintz RL, Butterfield G, Hoff-
Clin Endocrinol Metab 2002;87(10):4522–4527. man AR, et al. Effect of recombinant human growth hormone on
31. Hak AE, Witteman JC, de Jong FH, Geerlings MI, Hofman A, the muscle strength response to resistance exercise in elderly men.
Pols HA. Low levels of endogenous androgens increase the risk J Clin Endocrinol Metab 1994;79(5):1361–1366.
of atherosclerosis in elderly men: the Rotterdam study. J Clin 51. Brill KT, Weltman AL, Gentili A, Patrie JT, Fryburg DA, Hanks
Endocrinol Metab 2002;87(8):3632–3639. JB, et al. Single and combined effects of growth hormone and
32. Isidori AM, Giannetta E, Greco EA, Gianfrilli D, Bonifacio V, testosterone administration on measures of body composition,
Isidori A, et al. Effects of testosterone on body composition, bone physical performance, mood, sexual function, bone turnover, and
metabolism and serum lipid profile in middle-aged men: a meta- muscle gene expression in healthy older men. J Clin Endocrinol
analysis. Clin Endocrinol (Oxf) 2005;63(3):280–293. Metab 2002;87(12):5649–5657.
33. Tenover JL. Testosterone and the aging male. J Androl 52. Giannoulis MG, Sonksen PH, Umpleby M, Breen L, Pentecost C,
1997;18(2):103–106. Whyte M, et al. The effects of growth hormone and/or testosterone
34. Corpas E, Harman SM, Blackman MR. Human growth hormone in healthy elderly men: a randomized controlled trial. J Clin
and human aging. Endocr Rev 1993;14(1):20–39. Endocrinol Metab 2006;91(2):477–484.
35. Brook CG, Hindmarsh PC. The somatotropic axis in puberty. 53. Mariotti S, Franceschi C, Cossarizza A, Pinchera A. The aging
Endocrinol Metab Clin North Am 1992;21(4):767–782. thyroid. Endocr Rev 1995;16(6):686–715.
36. Veldhuis JD, Iranmanesh A, Bowers CY. Joint mechanisms of 54. Oddie TH, Meade JH Jr, Fisher DA. An analysis of published data
impaired growth-hormone pulse renewal in aging men. J Clin on thyroxine turnover in human subjects. J Clin Endocrinol Metab
Endocrinol Metab 2005;9(7):4177–4183. 1966;26(4):425–436.
37. Sonntag WE, Forman LJ, Miki N, Trapp JM, Gottschall PE, 55. Olsen T, Laurberg P, Weeke J. Low serum triiodothyronine and
Meites J. L-Dopa restores amplitude of growth hormone pulses high serum reverse triiodothyronine in old age: an effect of disease
in old male rats to that observed in young male rats. not age. J Clin Endocrinol Metab 1978;47(5):1111–1115.
Neuroendocrinology 1982;34(3):163–168. 56. Sawin CT, Geller A, Kaplan MM, Bacharach P, Wilson PW,
38. Takahashi S, Gottschall PE, Quigley KL, Goya RG, Meites J. Hershman JM. Low serum thyrotropin (thyroid-stimulating
Growth hormone secretory patterns in young, middle-aged and hormone) in older persons without hyperthyroidism. Arch Intern
old female rats. Neuroendocrinology 1987;46(2):137–142. Med 1991;151(1):165–168.

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
180 HS Chahal et al

57. Sawin CT, Geller A, Wolf PA, Belanger AJ, Baker E, 72. Greendale GA, Unger JB, Rowe JW, Seeman TE. The relation
Bacharach P, et al. Low serum thyrotropin concentrations as a between cortisol excretion and fractures in healthy older
risk factor for atrial fibrillation in older persons. N Engl J Med people: results from the MacArthur studies. J Am Geriatr Soc
1994;331(19):1249–1252. 1999;47(7):799–803.
58. Mariotti S, Barbesino G, Caturegli P, Bartalena L, Sansoni P, 73. Purnell JQ, Brandon DD, Isabelle LM, Loriaux DL, Samuels MH.
Fagnoni F, et al. Complex alteration of thyroid function in healthy Association of 24-hour cortisol production rates, cortisol-binding
centenarians. J Clin Endocrinol Metab 1993;77(5):1130–1134. globulin, and plasma-free cortisol levels with body composition,
59. Cuttelod S, Lemarchand-Beraud T, Magnenat P, Perret C, Poli S, leptin levels, and aging in adult men and women. J Clin Endocrinol
Vannotti A. Effect of age and role of kidneys and liver on Metab 2004;89(1):281–287.
thyrotropin turnover in man. Metabolism 1974;23(2):101–113. 74. Yen SS, Laughlin GA. Aging and the adrenal cortex. Exp Gerontol
60. Greenspan SL, Klibanski A, Rowe JW, Elahi D. Age-related 1998;33(7–8):897–910.
alterations in pulsatile secretion of TSH: role of dopaminergic 75. Baulieu EE. Dehydroepiandrosterone (DHEA): a fountain of
regulation. Am J Physiol 1991;260(3 Pt 1):E486–491. youth? J Clin Endocrinol Metab 1996;81(9):3147–3157.
61. Chiovato L, Mariotti S, Pinchera A. Thyroid diseases in the 76. Orentreich N, Brind JL, Rizer RL, Vogelman JH. Age changes
elderly. Baillières Clin Endocrinol Metab 1997;11(2):251–270. and sex differences in serum dehydroepiandrosterone sulfate
62. Pinchera A, Mariotti S, Barbesino G, Bechi R, Sansoni P, Fagi- concentrations throughout adulthood. J Clin Endocrinol Metab
olo U, et al. Thyroid autoimmunity and ageing. Horm Res 1984;59(3):551–555.
1995;43(1–3):64–68. 77. Labrie F, Belanger A, Cusan L, Gomez JL, Candas B. Marked
63. Bergendahl M, Iranmanesh A, Mulligan T, Veldhuis JD. Impact decline in serum concentrations of adrenal C19 sex steroid
of age on cortisol secretory dynamics basally and as precursors and conjugated androgen metabolites during aging.
driven by nutrient-withdrawal stress. J Clin Endocrinol Metab J Clin Endocrinol Metab 1997;82(8):2396–2402.
2000;85(6):2203–2214. 78. Vermeulen A. Dehydroepiandrosterone sulfate and aging. Ann N Y
64. Parker CR Jr, Slayden SM, Azziz R, Crabbe SL, Hines GA, Acad Sci 1995;774:121–127.
Boots LR, et al. Effects of aging on adrenal function in the human: 79. Parker CR Jr, Mixon RL, Brissie RM, Grizzle WE. Aging alters
responsiveness and sensitivity of adrenal androgens and cortisol zonation in the adrenal cortex of men. J Clin Endocrinol Metab
to adrenocorticotropin in premenopausal and postmenopausal 1997;82(11):3898–3901.
women. J Clin Endocrinol Metab 2000;85(1):48–54. 80. Allolio B, Arlt W. DHEA treatment: myth or reality? Trends
65. Waltman C, Blackman MR, Chrousos GP, Riemann C, Har- Endocrinol Metab 2002;13(7):288–294.
man SM. Spontaneous and glucocorticoid-inhibited adrenocorti- 81. Gurnell EM, Chatterjee VK. Dehydroepiandrosterone replacement
cotropic hormone and cortisol secretion are similar in healthy therapy. Eur J Endocrinol 2001;145(2):103–106.
young and old men. J Clin Endocrinol Metab 1991;73(3):495–502. 82. Morales AJ, Nolan JJ, Nelson JC, Yen SS. Effects of replacement
66. Van CE, Leproult R, Kupfer DJ. Effects of gender and age on dose of dehydroepiandrosterone in men and women of advancing
the levels and circadian rhythmicity of plasma cortisol. J Clin age. J Clin Endocrinol Metab 1994;78(6):1360–1367.
Endocrinol Metab 1996;81(7):2468–2473. 83. Baulieu EE, Thomas G, Legrain S, Lahlou N, Roger M, Debuire B,
67. Sherman B, Wysham C, Pfohl B. Age-related changes in the et al. Dehydroepiandrosterone (DHEA), DHEA sulfate, and aging:
circadian rhythm of plasma cortisol in man. J Clin Endocrinol contribution of the DHEAge Study to a sociobiomedical issue.
Metab 1985;61(3):439–443. Proc Natl Acad Sci USA 2000;97(8):4279–4284.
68. Laughlin GA, Barrett-Connor E. Sexual dimorphism in the 84. Wolf OT, Neumann O, Hellhammer DH, Geiben AC, Stras-
influence of advanced aging on adrenal hormone lev- burger CJ, Dressendorfer RA, et al. Effects of a two-week physi-
els: the Rancho Bernardo Study. J Clin Endocrinol Metab ological dehydroepiandrosterone substitution on cognitive perfor-
2000;85(10):3561–3568. mance and well-being in healthy elderly women and men. J Clin
69. Seeman TE, Robbins RJ. Aging and hypothalamic–pituitary– Endocrinol Metab 1997;82(7):2363–2367.
adrenal response to challenge in humans. Endocr Rev 85. Arlt W, Callies F, Koehler I, van Vlijmen JC, Fassnacht M,
1994;15(2):233–260. Strasburger CJ, et al. Dehydroepiandrosterone supplementation in
70. Seeman TE, McEwen BS, Singer BH, Albert MS, Rowe JW. healthy men with an age-related decline of dehydroepiandrosterone
Increase in urinary cortisol excretion and memory declines: secretion. J Clin Endocrinol Metab 2001;86(10):4686–4692.
MacArthur studies of successful aging. J Clin Endocrinol Metab 86. Flynn MA, Weaver-Osterholtz D, Sharpe-Timms KL, Allen S,
1997;82(8):2458–2465. Krause G. Dehydroepiandrosterone replacement in aging humans.
71. Dennison E, Hindmarsh P, Fall C, Kellingray S, Barker D, J Clin Endocrinol Metab 1999;84(5):1527–1533.
Phillips D, et al. Profiles of endogenous circulating cortisol and
bone mineral density in healthy elderly men. J Clin Endocrinol
Metab 1999;84(9):3058–3063.

J Pathol 2007; 211: 173–180 DOI: 10.1002/path


Copyright  2007 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

You might also like