You are on page 1of 20

Chapter

Regulation of Growth Hormone Secretion

8
Linear growth in humans is dependent on a complex pituitary where they control the secretion of GH.
interplay of various endocrine factors including: Figure 8.1 illustrates the homeostatic feedback path-
growth hormone (GH), insulin-like growth factors ways involving GH, hypothalamic neuropeptides
(e.g., IGF-1), ghrelin, thyroid hormones, glucocorti- (GHRH and SOM), ghrelin and IGF-1 from the
coids (GCs), sex hormones and non-endocrine mod- liver. Ghrelin is the most potent GH secretagogue
ulators such as nutritional status, familial growth known. Ghrelin infusion, in men and women, syner-
pattern, physical and psychosocial health. The pri- gizes with GHRH to stimulate pulsatile release of GH
mary determinants, however, are GH, GH-stimulated from somatotrophs, but likely exerts a positive feed-
IGF-1 production by the liver, plus ghrelin derived back on GHRH neurons as well (Veldhuis et al., 2008).
from the stomach. Ghrelin also plays a major role in In contrast, IGF-1 inhibits GHRH release but stimu-
the control of food intake (see Chapter 4). Its involve- lates SOM release, a combination that attenuates GH
ment in GH secretion is covered in Section 8.1. secretion. Note that GH also controls its own release
This chapter will focus on the regulation of pitui- via autocrine regulation exerted via GH receptors on
tary GH secretion and the pathophysiological impli- somatotrophs (Figure 8.1).
cations of abnormal GH production. GH is secreted
from anterior pituitary somatotrophs, which make up 8.2 Pulsatile Secretion
approximately 50% of the cell population of the gland
Similar to various other pituitary hormones, GH is
(Heaney and Melmed, 2004). At least five different
secreted in an episodic manner. Typical GH secretion
isoforms of GH exist, encoded by homologous genes,
consists of a minimal basal secretory rate where GH
Although only one, GH1, is expressed in somato-
levels drop to <0.04 mcg/L, interspersed by bursts or
trophs (Lim et al., 2014).
pulses. Although the average frequency of GH pulses
in both men and women is similar (approx. 11 over 24
8.1 Regulation of GH Secretion hours), the mean GH concentration is significantly
GH secretion is regulated through both central (GH- elevated in women (Veldhuis et al., 2011; Roelfsema
releasing hormone [GHRH] and somatostatin and Veldhuis, 2016). The pulse frequency is also
[SOM]) and peripheral (ghrelin and IGF-1) factors. higher in prepubertal children and adolescents com-
An overview of the orchestration of GH release is pared to adult levels (Steyn et al., 2016). Evidence
shown in Figure 8.1. The hypothalamic neuropeptide from animal experiments confirm that GHRH and
hormones – GHRH (stimulatory) and SOM (inhibi- SOM are released as pulses from the hypothalamus
tory) – constitute the principal central factors of GH into the hypophyseal blood circulation (Thomas et al.,
regulation. However, the hormone ghrelin – released 1991; Goldenberg and Barkan, 2007) and it is likely,
from the stomach – exerts a powerful stimulatory although unproven, that neuronal secretion of human
effect on GH secretion, whereas IGF-1, secreted GHRH and SOM is also inherently pulsatile.
from the liver, inhibits GH secretion. Cell bodies of However, the overall pituitary output of GH is
the GHRH and SOM neurons are located in close achieved, not by increasing pulse frequency, but by
proximity to each other in the basal infundibulum/ amplifying pulse, or burst, size. Pulse amplitude is
median eminence region of the hypothalamus regulated by a combination of GHRH stimulation,
(Proudan et al., 2015). Both peptides are released inhibition by SOM, plus a synergistic stimulation by
from nerve terminals and are transported through ghrelin. GH pulses are particularly evident at night
134 the hypophyseal portal system to reach the anterior (Figure 8.2; Van Cauter et al., 1998).

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Figure 8.1 Regulation of GH secretion. Pituitary secretion of GH is stimulated by pulsatile secretion of hypothalamic GHRH. The amplitude
of the GH pulse is modulated by an inhibitory effect of SOM. GH stimulates synthesis of IGF-1 in the liver; about 85% of circulating IGF-1 is liver-
derived. IGF-1 inhibits GH secretion at both hypothalamic and pituitary levels by reducing GHRH, and by stimulating SOM secretion. GH also
acts as an autocrine factor to control its own secretion from the pituitary. Ghrelin, released from the stomach, potently increases GH release by
boosting hypothalamic GHRH secretion. It also synergizes with GHRH to modulate GH pulse amplitude, but its overall role in physiological GH
regulation remains incompletely defined. Reproduced with permission (Kargi and Merriam, 2013). Abbreviations: GH, growth hormone; GHRH,
GH releasing hormone; IGF-1, insulin-like growth factor 1.

This figure shows that sleep deprivation abolishes the night when meals are not anticipated. It is thought
GH secretion and that daytime sleep, several hours that ghrelin may act as a positive modifier of GH
later, restores the sleep-related surge of GH. pulsatile secretion. In fact, intravenous boluses of
Nevertheless, despite the loss of the normal sleep- ghrelin induced slow-wave sleep (SWS) and a signifi-
induced peak of GH, daytime pulses of GH are cant increase in GH secretion in young men (see also
increased, so that the overall quantity of GH per 24 Section 8.3; Weikel et al., 2003).
hours is not compromised by sleep deprivation Clinical implications: Macimorelin is a ghrelin
(Brandenberger and Weibel, 2004). This is also con- receptor agonist that stimulates the production of
firmed in studies where subjects were deprived of sleep GH. This agent is used for testing of GH deficiency
for 24 hours; that is, the sleep-associated peak of GH (see later in this chapter).
was normal (Schüssler et al. 2006; see also Section 8.3). Clinical relevance: The pulsatile nature of the GH
A clear association of increases in ghrelin secretion secretory pattern has profound diagnostic and therapeu-
and GH surges is revealed when GH and ghrelin are tic implications. Not only does a single random measure-
assayed in plasma from a group of fed young men ment of GH provide limited information on GH levels,
(Figure 8.3; Nass et al., 2011). The meal-associated but targeting medical therapies in GH disorders also
peaks of ghrelin are coincident with those of GH. In cannot be reliably based on GH levels alone
addition, ghrelin and GH peaks are coincident during (Goldenberg and Barkan, 2007). 135

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Figure 8.2 Sleep-induced GH secretion and effect of sleep deprivation. Plasma GH in ten normal young men studied during a 52-hour period
including 8 hours of normal, nocturnal sleep, 28 hours of sleep deprivation and 8 hours of daytime sleep. GH secretion is abolished by sleep
deprivation, but is reinstated by a sleep period during the normal daytime. Figure is based on data obtained from Van Cauter et al. (1998).

Lunch

70
100

60
Dinner
; pg/mL)

Bkfst
50 10 GH (
; microgm/L)

40
Acyl-Ghrelin (

30 1

20

0.1
10

sleep
0
0800 1000 1200 1400 1600 1800 2000 2200 2400 0200 0400 0600 0800 1000
Clock time (hour)
Figure 8.3 Coincidence of ghrelin and GH secretion in the fed state. The figure illustrates that the meal-associated peaks of ghrelin are
coincident with those of GH. Ghrelin and GH peaks are also coincident during the night, when meals are not anticipated, emphasizing that
ghrelin may act as a modifier of GH secretion. Values are mean+/-SEM plasma acyl-ghrelin (picograms per milliliter) and serum GH levels
(micrograms per liter, shown on log scale); n=8 young men during the fed state. Reproduced with permission (Nass et al., 2011).

8.3 Other Factors Regulating GH whereas others are extrinsic, such as age, gender, sex
hormones, glucose, amino acids, sleep, feeding, exer-
Secretion cise, stress, illness, obesity and medication (Table 8.1;
In addition to GHRH, SOM and ghrelin, GH secre- Fanciulli et al., 2009). The effects of some of these
tion is sensitive to an array of other influences. Some regulatory factors form the underlying basis of clinical
136 of these are best described as neural (see Section 8.4), GH testing.

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Table 8.1 Regulators of GH secretion in humans. Reproduced with permission (Fanciulli et al., 2009).

Effector Increases GH Reduces GH Notes


Alpha1-adrenergic receptors ✔
Alpha2-adrenergic receptors ✔
Beta-adrenergic receptors ✔
Amino acids ✔
Anorexia nervosa ✔
Bombesin ✔
Dopamine ✔
Muscarinic receptors ✔
Cortisol ✔ ✔ Dose dependent
Diabetes mellitus ✔
Estrogen ✔ Increases pulse
amplitude
Exercise ✔
Free fatty acids ✔
γ-aminobutyric acid ✔
Galanin ✔
Ghrelin ✔
GHRH ✔
GH-releasing peptide ✔
Glucose ✔
Histamine ✔
Hypoglycemia ✔
Hypothyroidism ✔
IGF-1 ✔
Leptin Inversely correlated with
GH levels
Neuropeptide Y ✔
Obesity ✔
Opiates ✔
Senescence ✔
Serotonin ✔
Starvation ✔
Stress ✔
Testosterone ✔
Thyrotropin ✔ Only in acromegaly

Gender and Sex Hormones In addition, detailed studies by Veldhuis et al


Young women have significantly higher blood levels (2011) revealed that total GH secretion per day, and
of GH throughout the day compared to young men the amount of GH released per pulse, was significantly
(Goldenberg and Barkan, 2007; Figure 8.4). However, higher in healthy adult women compared to men,
this is not the case in older men and post-menopausal whereas the number of pulses per 24 hours was not
women, especially during sleep when secretory epi- different. During the menstrual cycle, when sex hor-
mone levels fluctuate (see Figure 3.1), there is a 137
sodes in women disappear (Latta et al., 2005).
Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

12 Figure 8.4 Women have higher GH levels than men. Sexual


dimorphism of GH secretion illustrated with data from a
young man (top) and a young woman (bottom). The
10 difference in secretory pattern is seen during sleep, but is
sleep also readily apparent in baseline states where larger
amplitude pulses of GH can be observed in the young
Plasma GH (ng/mL)

8 woman. Reproduced with permission (Goldenberg and


Barkan, 2007) and redrawn.

0
0600 noon 1800 2400 0600
14
sleep
12

10
Plasma GH (ng/mL)

0
0600 noon 1800 2400 0600
Clock time (hour)

significant increase in GH secretion during the luteal relationship holds for men, but is unconvincing for
phase, suggesting that progesterone, and possibly women. For example, testosterone supplementation
estradiol, might be a stimulatory factor for GH secre- in healthy men drives secretion of GH and IGF-1
tion in normal cycling women (Caufriez et al., 2009). (Veldhuis et al., 2005), although this action of testos-
GH levels typically fall in post-menopausal women, terone is mediated by its conversion to estradiol (aro-
and secretion can be restored by estrogen treatment matization) in the brain (Weissberger and Ho, 1993;
(Kalleinen et al., 2008). These data indicate a role for Veldhuis et al., 1997). The mechanism by which estra-
sex hormones in regulating GH secretion. This is also diol regulates GH secretion in women is incompletely
evident in children, where there is strong evidence for understood. One possible explanation for estrogen-
sex hormone stimulation of GH in boys and girls induced GH release is that estrogen leads to reduced
during puberty (Meinhardt and Ho, 2006). For exam- hepatic production of IGF-1, which in turn exerts a
ple, GH secretion is restored in hypogonadal boys stimulatory effect on GH release; that is, estradiol
treated with testosterone (Veldhuis et al., 1997; attenuates the IGF-1-mediated negative feedback,
138 Giustina et al., 1997). However, in adults, the thereby increasing GH release. Another possibility is

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Figure 8.5 Increases in SWS coincident with GH secretion. (A) Profiles of sleep stages, (B) slow-wave activity (EEG spectral power in the 0.5–4 Hz
frequency range), (C) plasma GH concentrations and (D) GH secretory rates derived from plasma GH levels in a healthy young man. Note the
temporal coincidence between major episodes of slow-wave activity and GH secretory pulses. Reproduced with permission (Van Cauter et al.,
2004) and redrawn.

an enhanced stimulatory effect of ghrelin on GH levels young men, as well as narcolepsy patients, with GHB,
in the presence of estradiol (Norman et al., 2014a) or a significantly increased SWS and GH secretion (Van
synergistic influence of estradiol on GHRH stimula- Cauter et al., 1997; Donjacour et al., 2011). The neural
tion of GH release (Norman et al., 2014b). In sum- control underlying the association of SWS and GH is
mary, although there are pronounced sex differences unknown, although the principal factor driving noc-
in GH secretion, the precise role, and mechanism of turnal GH secretion is GHRH; that is, an overnight
action, of gonadal hormones in men and women infusion of a GHRH antagonist largely eliminated GH
remains unclear. release (Ocampo-Lim et al., 1996). This suggests that
Clinical implications: The IGF-1 lowering effect of GHB increases the release of endogenous GHRH.
estrogen has led to the use of estrogen as an additional
therapeutic option in patients with excessive GH pro- Age
duction (acromegaly). GH is present in human fetal pituitary from 8 weeks of
gestation and is measurable in fetal serum from 12
Sleep weeks (Kaplan et al., 1976). Pulsatile secretion of GH
GH secretion is preferentially increased during is detectable in plasma immediately after birth, but
human sleep (Figures 8.2 and 8.3) and is closely asso- declines thereafter (Miller et al., 1993). GH levels are
ciated with slow wave sleep (SWS) (Figure 8.5; van higher than normal in preterm infants, leading to the
Cauter et al., 2004). suggestion that this might be responsible for neuro-
One of the few drugs known to increase SWS is γ- cognitive deficits seen later in life (Scratch et al.,
hydroxybutyrate (GHB), and treatment of healthy 2015). In normal children, however, GH levels 139

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

A deficiency of GH, either congenital or acquired, is


associated with distinct clinical outcomes. Childhood
onset GH deficiency (GHD) typically leads to short
stature, growth failure and delayed bone age, whereas
adult GHD is associated with loss of muscle mass,
increased adiposity, poor exercise capacity and dete-
rioration of mental function (Sattler, 2013; Bartke et al.,
2016; Milman et al., 2016). Although data on mortality
associated with GHD in humans are conflicting, mice
with mutational defects in somatotropic (GH/IGF-1)
action – and therefore with compromised growth and
body size – have remarkably extended lifespan, with
retention of physical and cognitive function and a
reduced incidence of age-related disease. Human stu-
dies on this phenomenon are limited (Milman et al.,
2016), although one large-scale investigation into
dwarf patients with mutations in the GHR (GH resis-
tance) revealed a highly significant reduction in the
incidence of cancer and diabetes compared to that in
normal relatives, although there was no observed
change in longevity (Guevara-Aguirre et al., 2011;
Guevara-Aguirre and Rosenbloom, 2015). In sum-
mary, age-related reductions in GH are regarded as
deleterious to human health and GH replacement as
an anti-aging therapy has been proposed (Kargi and
Merriam, 2013; Sattler, 2013). However, other evidence
indicates that GH replacement in adults may induce
unwanted consequences, such as increased insulin
Figure 8.6 Age-dependent decrease in GH secretion during 24 hours.
Mean (+/-SEM) values for GH for each age group plotted as total GH resistance and possibly cancer.
release in 24 hours (top) and as GH during sleep only (bottom). Clinical implications: Smaller doses of GH replace-
Probability levels, by analysis of variance, revealed a significant effect ment are typically required in the elderly based on the
of age in both total secretion (p<0.02) and in sleep-related GH
(p<0.001). Data were grouped by age as follows: 16–25 years (n=42); natural decline of GH with age. Section 8.8 will
26–35 years (n=28); 36–50 years (n=26); 51–60 years (n=23); 61–70 describe an example of clinical GHD.
years (n=18); 71–83 years (n=12). Figure is based on data obtained
from Van Cauter et al. (2000).
Glucose
increase substantially through the process of puberty, Glucose administration has a profound effect on GH
corresponding with the period of maximum height secretion. Typically, glucose administration in normal
velocity. In adolescents, the mass of GH per GH pulse individuals is associated with a biphasic GH response –
increases by up to ten-fold (Christoforidis et al., 2005; an initial SOM-mediated GH suppression is followed,
Veldhuis et al., 2006). These pubertal increases in GH 3–5 hours later, by an increase in GH release. On the
output are driven by aromatization of testosterone to other hand, hypoglycemia leads to release of GH, in
estradiol in boys and by estradiol in girls (Veldhuis tandem with hormones such as cortisol, via a reduction
et al., 1997). After puberty, GH secretion declines in SOM control (Giustina and Veldhuis, 1998). The
steadily to reach very low levels as early as 50–60 sensitivity of GH levels to glucose constitutes the basis
years. The decline is observable as an attenuation of of clinical testing of GH disorders. For instance, failure
pulse amplitude (Blackman, 2000) as well as total of GH to suppress after oral glucose administration is
secretion in 24 hours. This is especially notable during regarded as the gold standard for the diagnosis of acro-
sleep (Figure 8.6; van Cauter et al., 2000; Junnila et al., megaly (excessive GH production), whereas failure of
2013) where the day–night rhythm largely disappears GH to rise after insulin-induced hypoglycemia suggests
140
(Latta et al., 2005; Chertman et al., 2015). GHD.
Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Clinical implications: Acromegaly is confirmed by patients – the influence of GC on GH secretion is in


measuring GH after administering 75 g oral glucose. fact biphasic; the biological activity of GC is depen-
GH levels suppress to <1 mcg/L (<0.4 mcg/L with sen- dent on dose and time of exposure. For example, in
sitive GH assay) in normal individuals. Failure to sup- hypocortisolemic patients, physiological doses of GC
press GH suggests acromegaly. GHD is confirmed by will restore GH secretion (Barkan et al., 2000). In this
measuring GH after an insulin-induced hypoglycemia study, patients with Addison’s disease were with-
of <2.2 mmol/L. GH levels typically rise to >5.0 mcg/L in drawn (24 hours) from cortisol treatment and then
normal individuals. Failure of GH to rise to >5.0 mcg/L given replacement doses of hydrocortisone to main-
suggests GHD. tain physiological levels of cortisol. Withdrawal of GC
maintenance impaired spontaneous GH secretion
Glucocorticoids (Mazziotti and Giustina, 2013), but GH levels were
As described in Chapter 5, the GC cortisol is a major increased significantly following replacement cortisol
influence on most cells of the body. However, negative infusion due entirely to a doubling of pulse amplitude
effects of GCs are well described and include impaired (Figure 8.7). Patients with idiopathic adrenocortico-
growth in childhood, and altered body composition and tropin (ACTH) deficiency are also deficient in GH,
bone metabolism in adults and children. The common and respond to cortisol with increases in GH levels as
view is that an excess of GCs, either endogenous or described for Addison’s patients (Giustina et al.,
exogenous, suppresses GH secretion. This inhibition 1989). The authors concluded that cortisol stimulates
might be imposed at several sites, including inhibition GH output by reducing the inhibitory influence of
of GH production and secretion, inhibition of GHRH hypothalamic SOM.
and ghrelin action and stimulation of SOM activity (see
Figure 8.1). Inhibitory Effects of GC on GH
Both short term (1 hour) and long term (> 3 months)
Stimulatory Effects of GC on GH treatment with pharmacological doses of GC reduce
Notwithstanding the view that GCs are inhibitory to GH secretion and inhibit the stimulatory effect of
GH secretion – for example, in Cushing’s syndrome GHRH on pituitary GH release (Mazziotti and

Figure 8.7 Cortisol-induced secretion of GH in patients with primary adrenal insufficiency (Addison’s disease). Figure shows data from two
patients (A and B) with long-standing Addison’s disease. Patients had received regular hydrocortisone replacement but were withdrawn from
treatment 24 hours prior to admission. Patients were then infused with either saline or hydrocortisone (19 mg/day) given in a pattern designed
to mimic the normal circadian rhythm in cortisol secretion (i.e., peaks in the early morning). Compared to saline infusions, cortisol induced a 141
significant increase in GH secretion. The graphs are based on data obtained from Barkan et al. (2000).
Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Figure 8.8 Biphasic effects of GCs on GH secretion. The figure outlines a model of the effect of circulating GCs on human GH secretion. The
ascending part of the curve (light blue) reveals the stimulatory effect of GCs observable in a GC-deficient state. At higher doses, the
well-described inhibition of GH becomes apparent. Data were extrapolated from studies in humans performed separately in patients with
hypoadrenalism and in those exposed to an excess of GCs, either endogenous or exogenous. Reproduced with permission (Mazziotti and
Giustina, 2013).

Giustina, 2013). Such effects are seen in patients with focused on drug-induced changes, implicate several
hypercortisolism (Cushing’s syndrome) and in those receptor types in controlling human GH secretion
undergoing long-term immunosuppressive GC treat- (Table 8.1). For example, clonidine and the anti-
ment, for example, following organ transplants. In depressant reboxetine act as hypothalamic α2-adre-
summary, GCs represent a critical component of the nergic stimuli for GH secretion (Schüle et al., 2004;
normal regulation of GH secretion. GC have both Steyn et al., 2016). Baclofen, a γ-aminobutyric acidB
stimulatory and inhibitory effects on GH secretion, agonist, also stimulates GH secretion in women
dependent on dose and time of exposure (Figure 8.8). (Orio et al., 2001). For serotonin, the evidence is
less convincing. Increased secretion of GH, follow-
ing insulin-induced hypoglycemia, is prevented by
8.4 Neural Control of GH Secretion the serotonin receptor antagonist cyproheptadine
Figure 8.1 illustrates that pituitary GH secretion is (Bivens et al., 1973), but the selective serotonin
primarily controlled by GHRH, SOM and ghrelin. reuptake inhibitor citalopram, a drug designed to
However, there are multiple stimulatory and inhibi- increase synaptic serotonin, significantly increased
tory signals, some described in Section 8.1, and ACTH secretion but had no effect on GH levels
some of them of brain origin, that impose further (Berardelli et al., 2010). On the other hand, acetyl-
levels of regulation (see Table 8.1; Fanciulli et al., choline has a profound stimulatory effect on GH
2009). Animal experiments have provided detailed secretion in normal men and women. Inhibition of
knowledge of the neural control systems (Steyn central acetylcholinesterase, by physostigmine, or by
et al., 2016), but evidence for such involvement in donepezil, markedly increased basal as well as
man is limited (Giustina and Veldhuis,1998; Steyn GHRH-stimulated GH secretion (Fedi et al., 2008;
et al., 2016). Extensive immunohistochemical stu- Obermayr et al., 2005). In addition, arginine- or
dies in human brain tissue revealed evidence for exercise-induced GH secretion was blocked by pre-
possible neurotransmitter/neuropeptide synaptic treatment with the cholinergic antagonist atropine
regulation of GHRH and/or SOM neurons, includ- (Casanueva et al., 1984). Inhibitory dopamine recep-
ing dopamine (inhibitory; Rotoli et al., 2011), neu- tors are thought to underlie the effects of the anti-
ropeptide Y (inhibitory; Deltondo et al., 2008), SOM psychotic olanzapine. In this study, olanzapine
(inhibitory; Proudan et al., 2015), enkephalin (sti- abolished the normal nocturnal secretion of GH in
mulatory; Olsen et al., 2014) and CRH (inhibitory; schizophrenic patients (Mann et al., 2006). Opioid
142 Peroski et al., 2016). Complementary reports, receptors appear to be stimulatory for human GH

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

secretion (Vuong et al., 2010). For example, a met- 8.5 Receptor Mechanisms that Control
enkephalin analog elevated GH secretion in young
men (Giusti et al., 1992), and morphine stimulated GH Secretion from Somatotrophs
GH release in acromegalic patients (Bhansali et al., As noted in Section 8.1, the principal factors
2005). In contrast, blockade of opioid receptors with that enable secretion of GH from somatotrophs
the antagonist naltrexone prevented GHRH induc- are GHRH, SOM and ghrelin, together with nega-
tion of GH release (Villa et al., 1997). In summary, tive feedback from IGF-1. Somatotrophs possess
the regulation of human GH secretion appears to be specific cell membrane receptors for each factor
under the control of the same neurotransmitters (Figure 8.9).
and neuropeptides already implicated in experimen- Three of them are of the GPCR class (described in
tal animals. It remains to be determined whether Chapter 1). Receptors for GHRH and ghrelin are
these influences are imposed on GHRH or on SOM stimulatory and are linked to cyclic adenosine mono-
neurons (or both). For example, opioids could phosphate (cAMP) and inositol triphosphate (IP3)
directly stimulate GHRH neurons or, alternatively, signals, respectively. GHRH and ghrelin act synergis-
SOM neurons could be inhibited by opioids to allow tically to induce intracellular calcium ion mobiliza-
GHRH secretion to occur. tion that, in turn, regulates GH secretion. In addition,

Figure 8.9 Receptor mechanisms


regulating GH secretion from
somatotrophs. Receptors for GHRH and
ghrelin are stimulatory G protein-
coupled, linked to cAMP and IP3
signals, respectively. GHRH and ghrelin
act synergistically to induce
intracellular calcium ion mobilization
that, in turn, regulates GH secretion.
These stimuli also induce GH gene
expression and GH synthesis in
somatotrophs. SOM binds to an
inhibitory G protein-coupled receptor
that reduces the accumulation of
cAMP, decreasing the release of GH
(Ben-Shlomo and Melmed, 2010).
Receptors for IGF-1 are inhibitory, but
not G protein-coupled, and consist of
four subunits: two alpha-subunits plus
two beta-subunits, held together by
covalent disulfide bonds (see Figure
8.10). Abbreviations: cAMP, cyclic
adenosine monophosphate; IP3,
inositol triphosphate; PLC,
phospholipase C; SOM, somatostatin.

143

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

These, in turn, activate intracellular signaling proteins


(insulin receptor substrate 1; IRS-1). Phosphorylated
IRS-1 then induces other signaling cascades that ulti-
mately regulate gene transcription. In the case of
somatotrophs this involves inhibition of GH synthesis
and release.
The human SOM receptor exists as five distinct
subtypes: SSTR1–SSTR5. All five subtypes are detect-
able in the human fetal pituitary, but adult pituitary
tissue contains only four of them (SSTR1, SSTR2,
SSTR3 and SSTR5). Of these, SSTR2 and SSTR5 are
most abundant on somatotrophs and are effective in
inhibiting secretion of GH. When activated simulta-
neously they induce a synergistic suppression of GH
release (Ren et al., 2003). Knowledge of these sub-
types has important implications for the therapeutic
use of synthetic SOM analogs, including the treat-
ment of neuroendocrine tumors (Theodoropoulou
and Stalla, 2013). SOM receptors are also found in
several human pituitary tumors; that is, GH-,
ACTH-, PRL- TSH-secreting adenomas and in
non-functioning pituitary tumors, suggesting that
these hormones may also be under inhibitory control
by SOM (Eigler and Ben-Shlomo, 2014). In addition,
SOM receptors are expressed in many peripheral
tissues, and influence secretion from the GI tract
(e.g., gastrin and secretin) and from the pancreas
(e.g., insulin and glucagon).
Figure 8.10 Schematic representation of the IGF-1 signaling system. Clinical relevance: Short- and long-acting SOM
IGF-1 receptors consist of four subunits: two alpha-subunits plus two
beta-subunits, held together by covalent disulfide bond. IGF- analogs such as octreotide, lanreotide and pasireo-
1 receptors possess intrinsic tyrosine kinase activity; that is, following tide are used for suppression of GH and normal-
IGF-1 binding, this kinase is activated, auto-phosphorylating ization of IGF-1 in acromegaly (Melmed, 2016). In
(activating) the beta-subunits. The activated beta-subunits then
phosphorylate intracellular signaling proteins (IRS-1). addition, these agents are also used for treatment of
Phosphorylated IRS-1 then induces other signaling cascades that TSH adenomas (see Chapter 10). Both octreotide
ultimately regulate gene transcription. In the case of somatotrophs and lanreotide have an affinity for SSTR2 and
this involves inhibition of GH synthesis and release.
SSTR5 whereas pasireotide has an affinity for
SSTR1, SSTR2, SSTR3 and SSTR5. Figure 8.11 illus-
these stimuli induce GH gene expression and GH trates the inhibitory effects of pasireotide treatment
synthesis in somatotrophs. SOM also binds to a on GH and IGF-1 levels in acromegalic patients
GPCR, but these receptors are inhibitory and prevent, (Schmid et al., 2016).
or reduce, the accumulation of cAMP, thus reducing
the release of GH (Ben-Shlomo and Melmed, 8.6 Receptor Mechanism for GH
2010; see Figure 8.9). Receptors for IGF-1 are not G Stimulation of IGF-1 Production in
protein-coupled and consist of four subunits: two
alpha-subunits plus two beta-subunits, held together Liver Cells
by covalent disulfide bonds, similar to the insulin Figure 8.1 reveals the liver as a major target for GH. In
receptor (Figure 8.10; Schwartz et al., 2000). IGF-1 contrast to the seven transmembrane domains of the
receptors (IGF-1 Rs) possess intrinsic tyrosine kinase GHRH receptor, the GHR has only a single trans-
activity; that is, following IGF-1 binding, this kinase is membrane domain. Binding of GH to the GHR
144 activated and auto-phosphorylates the beta subunits. induces the formation of a dimer (Figure 8.12).

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Figure 8.11 Inhibitory effect of


pasireotide on GH and IGF-1 secretion in
acromegalic patients. Patients were
treated with long-acting release
pasireotide (40 or 60 mg intramuscular,
every 28 days) for a period of 24 weeks.
Values of GH and IGF-1 in blood were
analyzed by immunoassay. Error bars
are presented as geometric means
(68% CI; n= numbers of patients in the
study at 24 weeks). Abbreviation: CON,
control. Data reproduced with the kind
permission of Dr. H. A. Schmid (Schmid
et al., 2016).

This type of receptor does not use cAMP as a in skeletal growth. Acromegaly, on the other hand,
second messenger but possesses docking sites for results from excessive GH production in postpubertal
janus kinases (JAKs), a family of tyrosine kinases individuals who have already achieved fusion of the
(see Chapter 1). Thus, when GH binds to its receptor, epiphyseal plates. These patients typically present
and following dimerization, JAK phosphorylates with growth of bone and soft tissue leading to enlar-
other intracellular chemical signals that act as second gement of hands, feet, skull and jaw; excessive sweat-
messengers. The most important of these is signal ing (due to the effect of GH on sweat glands);
transduction and activator of transcription (STAT). visceromegaly and obstructive sleep apnea due to
Once phosphorylated, STAT dimerizes and travels enlargement of tongue and soft tissue. If left untreated
into the cell nucleus where it modulates gene expres- patients with acromegaly die early of cardiovascular
sion for IGF-1 (Figure 8.12). Secretion of IGF-1 from or respiratory complications and cancer. The com-
the liver stimulates bone and muscle growth (Reiter monest etiology of gigantism and acromegaly is an
and Rosenfeld, 2008; Rozario et al., 2015). underlying GH-producing pituitary adenoma.
Clinical relevance: Acromegaly is characterized by Although mostly sporadic, these benign tumors can
somatotroph adenomas and hypersecretion of GH, also be familial in conditions such as multiple endo-
which leads to elevated IGF-1. Pegvisomant is a crine neoplasia type 1, familial isolated pituitary ade-
GHR antagonist that blocks the effect of GH at the noma, McCune–Albright syndrome and Carney
receptor and consequently reduces the production of complex. Acromegaly is suspected in patients with
IGF-1. Pegvisomant is used for treatment of acrome- typical clinical features and is confirmed by failure
galy (Franck et al., 2016; see Section 8.7). of GH to suppress to <1 mcg/L (or <0.4 mcg/L with
sensitive GH assay) after administration of a 75 g oral
glucose challenge. Serum IGF-1 is elevated in patients
8.7 GH Excess with excessive GH and is used as a marker of excessive
Two distinct clinical syndromes – gigantism and acro- GH activity.
megaly – are associated with excessive GH produc- The primary management of acromegaly is trans-
tion. Gigantism results from excessive prepubertal sphenoidal surgical excision of the tumor. Patients
GH secretion and is characterized by a rapid increase who are either unable to achieve a remission after 145

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

In uncontrolled female acromegaly patients, estrogen


therapy can also be added to these agents to further
reduce IGF-1 (Shimon and Barkan, 2012).
Occasionally, radiation therapy is also used in patients
with residual tumor that fails to respond to medical
therapy. The aim of therapy is removal/shrinkage of
the tumor and normalization of GH and IGF-1 levels.
Unfortunately, several complications related to acro-
megaly are irreversible despite achieving biochemical
normalization.

8.7.1 A Case of Gigantism


A 15-year-old male was referred to the endocrinol-
ogy clinic for excessive fatigue and sleepiness. His
birth weight was 3.5 kg, and he had normal growth
until age 11, after which he experienced an acceler-
ated increase in his height and weight. About 6
months ago he developed pain in his left hip, tin-
gling in both hands and noticed deteriorating vision
for 2 months prior to presentation. His mother
mentioned that he snored heavily during sleep and
he slept on a semi-reclined couch because he was
unable to lay flat. His paternal aunt had undergone
surgery for a pituitary tumor of unknown etiology
17 years ago. On examination, his height was
198.12 cm (mid-parental height=172.3 cm) and
weight was 126.2 kg (BMI=32.2 kg/m2). His shoe
size was 16EEE (extra-wide). He had evidence of
scoliosis and had bilateral field of vision constriction
on confrontation.
Endocrine investigations were as follows:
Cortisol=298 nmol/L (184–512)
Figure 8.12 GHR signaling in the production of IGF-1 by the liver. TSH=2.37 mIU/L (0.35–5.4)
The GHR, unlike those for GHRH and SOM, has only a single
transmembrane domain. Binding of GH to the GHR induces the Free thyroxine (fT4)=8.8 pmol/L (11–19)
formation of a dimer that possesses docking sites for janus kinases PRL=15.4 mcg/L (2.1–17.7)
(JAKs), a family of tyrosine kinases. Thus, following GH-induced GH=13.4 mcg/L (<3.0)
dimerization, JAK phosphorylates other intracellular chemical
signals that act as second messengers. The most important of IGF-1=1600 mcg/L (232–1077)
these is signal transduction and activator of transcription (STAT). Testosterone=<0.3 nmol/L (8.0–32)
Once phosphorylated, STAT dimerizes and travels into the cell Follicle stimulating hormone (FSH)=0.5 IU/L
nucleus where it modulates gene expression for IGF-1. IGF-1 is
then secreted from the liver, binds to a binding protein (BP) and is (1.5–9.3)
transported to target tissues such as bone and muscle. Luteinizing hormone (LH)=0.4 IU/L (1.4–18.1)
Abbreviations: BP, binding protein; mRNA, messenger ribonucleic Ca=2.31 mmol/L (2.23–2.58)
acid. Reproduced with permission (Junnila et al., 2013).
Nadir GH after an oral glucose tolerance test=7.9
mcg/L
surgery or cannot undergo surgery are treated with Figure 8.13A shows a magnetic resonance image
medical therapy. The currently available pharmacolo- (MRI) confirming the presence of a macroadenoma
gic options include: SOM analogs (octreotide, lanreo- and Figure 8.13B reveals that a Goldmann visual field
tide and pasireotide; see Figure 8.11), dopamine test showed bilateral visual field defects.
agonists (bromocriptine and cabergoline) and the A diagnosis of gigantism or early-onset acromegaly
146 GHR antagonist pegvisomant (Franck et al., 2016). was made on the basis of typical clinical features and

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

A. MRI reveals a macroadenoma

B. Goldmann visual field test shows bilateral visual field defects

Figure 8.13 (A) MRI scans confirming the presence of a macroadenoma; (B) Goldmann visual field test results showing bilateral visual field defects.

elevated serum IGF-1 and non-suppressibility of GH to 8.8 GH Deficiency


<0.4 mcg/L after oral glucose. Based on the family
GHD can occur as a result of congenital pituitary
history, invasive macroadenoma, male gender and
defects, pituitary tumors and infiltrative disorders,
young age at presentation he was offered genetic test-
pituitary surgery or radiation therapy and head
ing. The test confirmed an aryl-hydrocarbon interact-
trauma. GHD in children typically presents with
ing receptor protein mutation associated with familial
growth failure and short stature, whereas adults
isolated pituitary adenomas (Imran et al., 2018). The
show decreased muscle and bone mass, increased
patient underwent trans-sphenoidal excision of the
cardiovascular risk factors and poor exercise reserve.
pituitary tumor followed by SOM analog therapy to
GHD is confirmed by failure of GH to rise in
achieve biochemical remission. Figure 8.14 shows the
response to provocative stimuli such as insulin-
patient with his mother 3 years following treatment.
induced hypoglycemia, L-Dopa, arginine, glucagon 147

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

asked his family physician to rule out GH deficiency


(GHD) and was referred to the endocrine clinic.
His past history was unremarkable, apart from a
head injury a year ago when he fell off his mountain
bike and briefly lost consciousness. He was admitted
to a local hospital where he underwent computed
tomography scans and was observed for a few days
and then discharged.
His pituitary investigations were as follows:
Cortisol=311 nmol/L (184–512)
TSH=1.17 mIU/L (0.35–5.4)
fT4=14.3 pmol/L (11–19)
PRL=7.7 mcg/L (2.1–17.7)
GH=1.08 mcg/L (<3.0)
IGF-1=88 mcg/L (175–288)
Testosterone=15.4 nmol/L (8.0–32)
FSH=2.2 IU/L (1.5–9.3)
LH=1.9 IU/L (1.4–18.1)
Peak GH after insulin-induced hypoglycemia=2.3
mcg/L
A diagnosis of GHD was made on the basis of low
IGF-1, and failure of GH to rise >5 mcg/L after insu-
Figure 8.14 Acromegalic patient, with his mother, shown 3 years
lin-induced hypoglycemia. The most likely underly-
following trans-sphenoidal removal of pituitary macroadenoma. ing etiology was GHD due to head injury. GH-
Reproduced with permission. replacement therapy was initiated and serum IGF-1
was normalized after 3 months (212 mcg/L). The
and the ghrelin receptor agonist macimorelin. patient experienced significant improvement in exer-
Different cut-off levels of GH response to these tests cise capacity with complete resolution of the fatigue.
are reported in the literature. Typically, a failure of
GH to rise to >5 mcg/L in normal individuals after 8.9 Chapter Summary
insulin-induced hypoglycemia of <2.2 mmol/L, or This chapter outlines the mechanisms by which GH
>2.8 mcg/L after macimorelin, confirms GHD. secretion is regulated to maintain GH homeostasis and
Management of GHD is in the form of injectable in those states of pathological GH secretion, such as
GH replacement, which has been shown to improve acromegaly. Human GH secretion appears to be under
height in children as well as bone and muscle mass, the control of the same neurotransmitters and neuro-
exercise capacity and quality of life in adults (Ahmid peptides already implicated in experimental animals,
et al., 2016). The aim of therapy is to normalize serum although it remains unknown whether these influences
IGF-1. are imposed on GHRH or SOM neurons (or both). GH
Unfortunately, the abuse of GH is high among secretion from somatotrophs is regulated by opposing
professional athletes and bodybuilding enthusiasts hypothalamic releasing factors: GHRH is stimulatory
(Baumann, 2012). Chronic use of excessive doses of and SOM inhibitory. In addition, GH levels are subject
GH are known to induce some features of acromegaly. to peripheral feedback pathways from IGF-1 (negative)
and ghrelin (positive). Ghrelin is the most potent GH
8.8.1 A Case of growth hormone deficiency secretagogue known, and synergizes with GHRH to
A 34-year-old athletic male presented with increasing stimulate pulsatile release of GH from somatotrophs.
fatigue and inability to keep up with his running Somatotrophs possess specific cell membrane receptors
group for the past 6 months. He was becoming fru- for each factor. Similar to other pituitary hormones,
strated because his routine investigations through his GH is secreted in an episodic (pulsatile) manner, espe-
family physician failed to show any concerning cially during SWS. GH secretion is sensitive to an array
148 abnormality. After searching the subject online, he of other influences such as age, gender, sex hormones,

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

glucose, amino acids, feeding, exercise, stress, illness, a. Hypoglycemia


obesity and medication. For example, sex hormones b. Pasireotide
stimulate GH secretion in pubertal children, and tes- c. Somatostatin (SOM)
tosterone supplementation in men drives GH and IGF- d. Insulin-like growth factor 1 (IGF-1)
1 release, but the role of estradiol in women is unclear. e. Ghrelin
In normal children, GH levels increase substantially 2. Which of the following statements on GH are
through the process of puberty, corresponding with incorrect?
the period of maximum height velocity. After puberty,
a. Secretion is negatively regulated by a
GH secretion declines steadily to reach very low levels
hypothalamic releasing factor.
as early as 50–60 years, especially notable during sleep.
b. Blood levels are higher in children than in
A deficiency of GH, either congenital or acquired, is
older adults.
associated with distinct clinical outcomes. Childhood-
c. Pulses of GH occur during sleep.
onset GHD leads to short stature and delayed bone age,
d. GH modulates the secretion of IGF-1 from the
whereas adult GHD is associated with loss of muscle
liver.
mass, increased adiposity, poor exercise capacity and
e. SOM stimulates GH secretion.
deterioration of mental function. A case of clinical
GHD is provided. 3. A pituitary adenoma in an adult, secreting high
Glucose administration in normal individuals levels of GH, causes:
induces a biphasic GH response – an initial suppres- a. Gigantism
sion followed, 3–5 hours later, by an increase in GH b. Enlargement of the liver
release. Hypoglycemia leads to release of GH. The c. Elevated blood levels of glucose
sensitivity of GH levels to glucose constitutes the d. Reduced circulating levels of IGF-1
basis of clinical testing of GH disorders: failure of e. Acromegaly
GH to suppress after oral glucose administration is 4. Which of the following statements concerning GH
the gold standard for the diagnosis of acromegaly, are correct? Select all that apply.
whereas failure of GH to rise after insulin-induced
hypoglycemia suggests GHD. a. GH shows increased secretion during SWS
GCs are generally regarded as inhibitory to GH sleep.
secretion, but the GH response to GC is in fact bipha- b. The systemic actions of GH are mediated by
sic. Treatment with pharmacological doses of GC IGF-1.
reduces GH secretion and inhibits the stimulatory c. IGF-1 is synthesized mainly in the anterior
effect of GHRH on pituitary GH release. Such effects pituitary.
are seen in patients with hypercortisolism (Cushing’s d. GH is required for growth in stature from
syndrome) and in those undergoing long-term immu- birth to puberty.
nosuppressive GC treatment. e. An increase in serum SOM will inhibit GH
Two distinct clinical syndromes – gigantism and secretion.
acromegaly – are associated with excessive GH pro- 5. A 12-year-old boy presented with short stature
duction. Gigantism occurs due to excessive prepuber- and reduced height velocity over the past 6
tal GH secretion and is characterized by a rapid months. Which of the following factors suggest a
increase in skeletal growth. Acromegaly results from possibility of GH deficiency (GHD)?
excessive GH production in postpubertal individuals. a. Previous diagnosis of celiac disease
A case of gigantism is presented. GHD can occur as a b. Both parents are short
result of congenital pituitary defects, pituitary tumors c. Subnormal serum IGF-1 level
and infiltrative disorders, pituitary surgery or radia- d. Severe asthma requiring glucocorticoid
tion therapy and head trauma. A case of GHD due to therapy
head trauma is presented. e. Presence of 16 mm pituitary adenoma on a
magnetic resonance image
8.10 Review Questions 6. Which of the following therapeutic options are
1. Which of the following inhibit growth hormone used for management of acromegaly?
149
(GH) secretion from somatotrophs? a. Cabergoline
Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

b. Lanreotide Blum W F, Alherbish A, Alsagheir A et al. (2018). The


c. Pasireotide growth hormone–insulin-like growth factor-1 axis in the
d. Estrogen diagnosis and treatment of growth disorders. Endocr
Connect 7, R212–R222.
e. Pegvisomant
Butler A A & le Roith D. (2001). Control of growth by the
7. Which of the following statements regarding somatropic axis: growth hormone and the insulin-like
GHD are correct? growth factors have related and independent roles. Annu
a. The diagnosis of GHD is based on the failure Rev Physiol 63, 141–164.
of GH to rise after an oral glucose load. Eigler T & Ben-Shlomo A. (2014). Somatostatin system:
b. Patients typically present with short stature molecular mechanisms regulating anterior pituitary
and growth failure as children. hormones. J Mol Endocr 53, R1–R19.
c. Serum IGF-1 levels are typically elevated in Melmed S. (2016). New therapeutic agents for acromegaly.
GHD. Nat Revs Endocr 12, 90–98.
d. GHD is associated with poor exercise tolerance Milman S, Huffman D M & Barzilai N. (2016). The
and muscle loss. somatotropic axis in human aging: framework for the
current state of knowledge and future research. Cell Metab
e. Patients with a history of significant head 23, 980–989.
trauma have a higher risk of developing GHD.
Monson J P, Brooke A M & Akker S. (2015). Adult growth
8. Features of acromegaly include: hormone deficiency. www.ncbi.nlm.nih.gov/books/
a. Rapid height gain in patients with postpubertal NBK278982/
onset of disease Murray P G & Clayton P E. (2016). Disorders of growth
b. Sleep apnea hormone in childhood. www.endotext.org
c. Loss of sweating Murray P G, Higham C E & and Clayton P E. (2015). The
d. Enlargement of the tongue hypothalamo–GH axis: the past 60 years. J Endocr 226,
T123–T140.
e. Failure of GH to rise after 75 g oral glucose
test Ranke M B & Wit J M. (2018). Growth hormone – past,
present and future. Nat Rev Endocr 14, 285–300.
9. Which one of the following predisposes to
Rozario K, Lloyd C & Ryan F J. (2015). GH and IGF-1
acromegaly? physiology in childhood. www.endotext.org
a. Celiac disease Sherlock M, Woods C & Sheppard M C. (2011). Medical
b. Head trauma therapy in acromegaly. Nat Rev Endocr 7, 291–300.
c. Familial isolated pituitary adenoma Steyn FJ, Tolle V, Chen C & Epelbaum J. (2016).
d. Sleep apnea Neuroendocrine regulation of growth hormone secretion.
e. Young age Compr Physiol 6, 687–735.
10. Pasireotide is used in the treatment of acromegaly. van der Lely A J, Tschop M, Heiman M L & Ghigo E.
Which of the following statements accounts for its (2004). Biological, physiological, pathophysiological,
and pharmacological aspects of ghrelin. Endocr Rev 25,
effectiveness? 426–57.
a. Pasireotide inhibits GH secretion through a G Vilar L, Vilar C F, Lyra R, Lyra R & Naves L A. (2017).
protein-coupled receptor. Acromegaly: clinical features at diagnosis. Pituitary 20,
b. Pasireotide inhibits IGF-1 secretion. 22–32.
c. Pasireotide synergizes with ghrelin to induce Walters T D & Griffiths A M. (2009). Mechanisms of growth
GH secretion. impairment in pediatric Crohn’s disease. Nat Rev
d. Pasireotide blocks SOM receptors. Gastroenterol Hepatol 6, 513–523.
e. Pasireotide binds to SOM receptors (SSTR) 1 –This paper contains excellent figures and a description
and 2. of the basic physiology of the GH/IGF-1 system.

Further Reading References


Ahmid M, Perry C G, Ahmed S F & Shaikh M G. (2016). Ahmid M, Perry C G, Ahmed S F & Shaikh M G. (2016).
Growth hormone deficiency during young adulthood and Growth hormone deficiency during young adulthood and
the benefits of growth hormone replacement. Endocr the benefits of growth hormone replacement. Endocr
150 Connect 5, R1–R11.
Connect 5, R1–R11.
Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Barkan A L, DeMott-Friberg R & Samuels M H. (2000). narcolepsy patients and healthy controls. Am J Physiol
Growth hormone (GH) secretion in primary adrenal Endocr Metab 300, E1069–E1075.
insufficiency: effects of cortisol withdrawal and patterned
Eigler T & Ben-Shlomo A. (2014). Somatostatin system:
replacement on GH pulsatility and circadian rhythmicity.
molecular mechanisms regulating anterior pituitary
Pituitary 3, 175–179.
hormones. J Mol Endocr 53, R1–R19.
Bartke A, List E O & Kopchick J J. (2016). The somatotropic
Fanciulli G, Delitala A & Delitala G. (2009). Growth hormone,
axis and aging: benefits of endocrine defects. Growth
menopause and ageing: no definite evidence for ‘rejuvenation’
hormone IGF Res 27, 41–45.
with growth hormone. Hum Reprod Update 15, 341–358.
Baumann G P. (2012). Growth hormone doping in sports: a
critical review of use and detection strategies. Endocr Revs Fedi M, Bach L A, Berkovic S F, Willoughby J O, Scheffer I E
33, 155–186. & Reutens D C. (2008). Association of a nicotinic receptor
mutation with reduced height and blunted physostigmine-
Ben-Shlomo A & Melmed S. (2010). Pituitary somatostatin stimulated growth hormone release. J Clin Endocr Metab 93,
receptor signaling. Trends Endocr Metab 21, 123–133. 634–637.
Berardelli R, Margarito E, Ghiggia F et al. (2010). Franck S E, Muhammad A, van der Lely A J & Neggers S J.
Neuroendocrine effects of citalopram, a selective serotonin (2016).Combined treatment of somatostatin analogues with
re-uptake inhibitor, during lifespan in humans. J Endocrinol pegvisomant in acromegaly. Endocr 52, 206–213.
Invest 33, 657–662.
Giusti M, Delitala G, Marini G et al. (1992). The effect of a
Bhansali A, Velayutham P, Sialy R & Sethi B. (2005). Effect met-enkephalin analogue on growth hormone, prolactin,
of opiates on growth hormone secretion in acromegaly. gonadotropins, cortisol and thyroid stimulating hormone in
Horm Metab Res 37, 425–427 healthy elderly men. Acta Endocrinol 127, 205–209.
Bivens C H, Lebovitz H E & Feldman J M. (1973). Inhibition Giustina A & Veldhuis J D. (1998). Pathophysiology of the
of hypoglycemia-induced growth hormone secretion by the neuroregulation of growth hormone secretion in
serotonin antagonists cyproheptadine and methysergide. experimental animals and the human. Endocr Revs 19,
New Engl J Med 289, 236–239. 717–797.
Blackman M R. (2000). Age-related alterations in sleep Giustina A, Romanelli G, Candrina R & Giustina G. (1989).
quality and neuroendocrine function; interrelationships Growth hormone deficiency in patients with idiopathic
and implications. JAMA 284, 879–881. adrenocorticotropin deficiency resolves during
Brandenberger G & Weibel L. (2004). The 24-h growth glucocorticoid replacement. J Clin Endocr Metab 68,
hormone rhythm in men: sleep and circadian influences 120–124.
questioned. J Sleep Res 13, 251–255. Giustina A, Scalvini T, Tassi C et al. (1997). Maturation of
Casanueva F F, Villanueva L, Cabranes J A, Cabezas-Cerrato J the regulation of growth hormone secretion in young males
& Fernandez-Cruz A. (1984). Cholinergic mediation of with hypogonadotropic hypogonadism pharmacologically
growth hormone secretion elicited by arginine, clonidine, and exposed to progressive increments in serum testosterone. J
physical exercise in man. J Clin Endocr Metab 59, 526–530. Clin Endocr Metab 82, 1210–1219.
Caufriez A, Leproult R, L’Hermite-Baleriaux M, Moreno- Goldenberg N & Barkan A. (2007). Factors regulating
Reyes R & Copinschi G. (2009). A potential role of growth hormone secretion in humans. Endocr Metab Clin N
endogenous progesterone in modulation of GH, prolactin Amer 36, 37–55.
and thyrotrophin secretion during normal menstrual cycle. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre
Clin Endocr, 71, 535–542. M et al. (2011). Growth hormone receptor deficiency is
Chertman L S, Merriam G R & Kargi A Y. (2015). Growth associated with a major reduction in pro-aging signaling,
hormone in aging. www.endotext.org cancer, and diabetes in humans. Sci Transl Med 3, 70ra13.
Christoforidis A, Maniadaki I & Stanhope R. (2005). Guevara-Aguirre J & Rosenbloom A L. (2015). Obesity,
Growth hormone/insulin-like growth factor axis during diabetes and cancer: insight into the relationship from a
puberty. Ped Endocr Rev 1, 5–10. cohort with growth hormone receptor deficiency.
Diabetologia 58, 37–42.
Deltondo J, Por I, Hu W et al. (2008). Associations between
the human growth hormone-releasing hormone- and Heaney A P & Melmed S. (2004). Molecular targets in
neuropeptide-y-immunoreactive systems in the human pituitary tumours. Nature Revs Cancer 4, 285–295.
diencephalon: a possible morphological substrate of the Imran S A, Aldahmani K A, Penney L et al. (2018). Unusual
impact of stress on growth Neurosci 153, 1146–1152. AIP mutation and phenocopy in the family of a young
Donjacour C E H M, Aziz N A, Roelfsema F et al. (2011). patient with acromegalic gigantism. Endocr Diabetes Metab
Effect of sodium oxybate on growth hormone secretion in Case Rep 2018, 17–0092.
151

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Junnila R K, List E O, Berryman D E, Murrey J W & somatostatin in postmenopausal women. Eur J Endocr 170,
Kopchick J J. (2013). The GH/IGF-1 axis in ageing and 121–129.
longevity. Nat Rev Endocr 9, 366–376. Obermayr R P, Mayerhofer L, Knechtelsdorfer M et al.
Kalleinen N, Polo-Kantola P, Irjala K et al. (2008). 24-hour (2005). The age-related down-regulation of the growth
serum levels of growth hormone, prolactin, and cortisol in hormone/insulin-like growth factor-1 axis in the elderly
pre- and postmenopausal women: the effect of combined male is reversed considerably by donepezil, a drug for
estrogen and progestin treatment. J Clin Endocr Metab 93, Alzheimer’s disease. Exp Gerontol 40, 157–163.
1655–1661. Ocampo-Lim B, Guo W, DeMott-Friberg R, Barkan A L &
Kaplan S L, Grumbach M M & Aubert M L. (1976). The Jaffe C A. (1996). Nocturnal growth hormone (GH)
ontogenesis of pituitary hormones and hypothalamic secretion is eliminated by infusion of GH-releasing
factors in the human fetus: maturation of central nervous hormone antagonist. J Clin Endocr Metab 81, 4396–4399.
system regulation of anterior pituitary function. Rec Prog Olsen J, Peroski M, Kiczek M, Grignol G, Merchenthaler I &
Horm Res 32, 161–243. Dudas B. (2014). Intimate associations between the
Kargi A Y & Merriam G R. (2013). Diagnosis and treatment endogenous opiate systems and the growth hormone-
of growth hormone deficiency in adults. Nat Rev Endocr 9, releasing hormone system in the human hypothalamus.
335–345. Neurosci 258, 238–245.
Latta F, Leproult R, Tasali E et al. (2005). Sex differences in Orio F Jr., Palombab S, Colao A et al. (2001). Growth
nocturnal growth hormone and prolactin secretion in hormone secretion after baclofen administration in
healthy older adults: relationships with sleep EEG variables. different phases of the menstrual cycle in healthy women.
Sleep 28, 1519–1524. Horm Res 55, 131–136.
Lim C T, Grossman A & Khoo B. (2014). Normal physiology Peroski M, Proudan N, Grignol G, Merchenthaler I & Dudas
of ACTH and GH release in the hypothalamus and anterior B. (2016). Corticotropin-releasing hormone (CRH)-
pituitary in man. www.endotext.org immunoreactive (IR) axon varicosities target a subset of
Mann K, Rossbach W, Müller M J et al. (2006). Nocturnal growth hormone-releasing hormone (GHRH)-IR neurons
hormone profiles in patients with schizophrenia treated in the human hypothalamus. J Chem Neuroanat 78,
with olanzapine. Psychoneuroendocrinol 31, 256–264. 119–124.

Mazziotti G & Giustina A. (2013). Glucocorticoids and the Proudan N, Peroski M, Grignol G, Merchenthaler I &
regulation of growth hormone secretion. Trends Endocr Dudas B. (2015). Juxtapositions between the
Metab 9, 265–276. somatostatinergic and growth hormone-releasing hormone
(GHRH) neurons in the human hypothalamus. Neurosci
Meinhardt U J & Ho K K Y. (2006). Modulation of growth 297, 205–210.
hormone action by sex steroids. Clin Endocr 65, 413–422.
Reiter E O & Rosenfeld R G. (2008). Normal and aberrant
Melmed S. (2016). New therapeutic agents for acromegaly. growth. Williams Textbook of Endocrinology, 11th Edition
Nat Revs Endocr 12, 90–98. (New York: Saunders Elsevier), 849–968.
Miller J D, Esparza A, Wright N M et al. (1993). Ren S-G, Taylor J, Dong J, Yu R, Culler M D & Melmed S.
Spontaneous growth hormone release in term infants: (2003). Functional association of somatostatin receptor
changes during the first four days of life. J Clin Endocr subtypes 2 and 5 in inhibiting human growth hormone
Metab 76, 1058–1062. secretion. J Clin Endocr Metab 88, 4239–4245.
Milman S, Huffman D M & Barzilai N. (2016). The Roelfsema F & Veldhuis J D. (2016). Growth hormone
somatotropic axis in human aging: framework for the dynamics in healthy adults are related to age and sex and
current state of knowledge and future research. Cell Metab strongly dependent on body mass index. Neuroendocr 103,
23, 980–989. 335–344.
Nass R, Gaylinn B D & Thorner M O. (2011). The role of Rotoli G, Grignol G, Hu W, Merchenthaler I & Dudas B.
ghrelin in GH secretion and GH disorders. Mol Cell Endocr (2011). Catecholaminergic axonal varicosities appear to
340, 10–14. innervate growth hormone-releasing hormone-
Norman C, Rollene N, Weist S M et al. (2014a). Short-term immunoreactive neurons in the human hypothalamus: the
estradiol supplementation potentiates low-dose ghrelin possible morphological substrate of the stress-suppressed
action in the presence of GHRH or somatostatin in older growth. J Clin Endocr Metab 96, E1606–E1611.
women. J Clin Endocr Metab 99, E73–E80. Rozario K, Lloyd C & Ryan F J. (2015). GH and IGF-1
Norman C, Rollene N L, Erickson D, Miles J M, Bowers C Y physiology in childhood. www.endotext.org
& Veldhuis J D. (2014b). Estradiol regulates GH releasing- Sattler F R. (2013). Growth hormone in the aging male. Best
peptide’s interactions with GH-releasing hormone and Pract Res Clin Endocr Metab 27, 541–555.
152

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009
Chapter 8: Regulation of Growth Hormone Secretion

Schmid H A, Brue T, Colao A et al. (2016). Effect of Van Cauter E, Latta F, Nedeltcheva A et al. (2004).
pasireotide on glucose- and growth hormone-related Reciprocal interactions between the GH axis and sleep.
biomarkers in patients with inadequately controlled Growth Hormone & IGF Res 14, S10–S17.
acromegaly. Endocr 53, 210–219. Veldhuis J D, Metzger D L, Martha P M Jr. et al. (1997).
Schüle C, Baghai T, Schmidbauer S, Bidlingmaier M, Estrogen and testosterone, but not a nonaromatizable
Strasburger C J & Laakmann G. (2004). Reboxetine acutely androgen, direct network integration of the hypothalamo-
stimulates cortisol, ACTH, growth hormone and prolactin somatotrope (growth hormone)– insulin-like growth factor
secretion in healthy male subjects. Psychoneuroendocr 29, I axis in the human: evidence from pubertal
185–200. pathophysiology and sex-steroid hormone replacement. J
Schüssler P, Uhr M, Ising M et al. (2006). Nocturnal ghrelin, Clin Endocr Metab 82, 3414–3420.
ACTH, GH and cortisol secretion after sleep deprivation in Veldhuis J D, Keenan D M, Mielke K, Miles J M & Bowers C
humans. Psychoneuroendocr 31, 915–923. Y. (2005). Testosterone supplementation in healthy
Schwartz M W, Woods S C, Porte D, Jr., Seeley R J & Baskin older men drives GH and IGF-I secretion without
D G. (2000). Central nervous system control of food intake. potentiating peptidyl secretagogue efficacy. Europ J Endocr
Nature 404, 661–671. 153, 577–586.

Scratch S E, Anderson P J, Doyle L W et al. (2015). High Veldhuis J D, Roemmich J N, Richmond E J & Bowers C Y.
postnatal growth hormone levels are related to cognitive (2006). Somatotropic and gonadotropic axes linkages in
deficits in a group of children born very preterm. J Clin infancy, childhood, and the puberty-adult transition.
Endocr Metab 100, 2709–2717. Endocr Revs 27, 101–140.

Shimon I & Barkan A. (2012). Estrogen treatment for Veldhuis J D, Reynolds G A, Iranmanesh A & Bowers C Y.
acromegaly. Pituitary 15, 601–607. (2008). Twenty-four hour continuous ghrelin
infusion augments physiologically pulsatile,
Steyn F J, Tolle V, Chen C & Epelbaum J. (2016). nycthemeral, and entropic (feedback-regulated) modes of
Neuroendocrine regulation of growth hormone secretion. growth hormone secretion. J Clin Endocr Metab 93,
Compr Physiol 6, 687–735. 3597–3603.
Theodoropoulou M & Stalla G K. (2013). Somatostatin Veldhuis J D, Roelfsema F, Keenan D M & Pincus S. (2011).
receptors: from signaling to clinical practice. Front Gender, age, body mass index, and igf-i individually and
Neuroendocr 34, 228–252. jointly determine distinct GH dynamics: analyses in
Thomas G B, Cummins J T, Francis H, Sudbury A W, one hundred healthy adults. J Clin Endocr Metab 96, 115–
McCloud P I & Clarke I J. (1991). Effect of restricted feeding 121.
on the relationship between hypophysial portal Villa P, Valle D, De Marinis L et al. (1997). Influence of
concentrations of growth hormone (GH)-releasing factor chronic naltrexone treatment on growth hormone secretion
and somatostatin, and jugular concentrations of GH in in normal subjects. Eur J Endocrinol 137, 631–634.
ovariectomized ewes. Endocr 128, 1151–1158.
Vuong C, Van Uum S H M, O’Dell L E, Lutfy K & Friedman
Van Cauter E, Plat L, Scharf M B et al. (1997). Simultaneous T C. (2010). The Effects of opioids and opioid analogs on
stimulation of slow-wave sleep and growth hormone animal and human endocrine systems. Endocr Rev 31, 98–
secretion by gamma-hydroxybutyrate in normal young 132.
men. J Clin Invest 100, 745–753.
Weikel J C, Wichniak A, Ising M et al. (2003). Ghrelin
Van Cauter E, Plat L & Copinschi G. (1998). Interrelations promotes slow-wave sleep in humans. Am J Physiol Endocr
between sleep and the somatotropic axis. Sleep 21, 553–566. Metab 284, E407–E415.
Van Cauter E, Leproult R & Plat L. (2000). Age-related Weissberger A J & Ho K K. (1993). Activation of the
changes in slow wave sleep and REM sleep and relationship somatotropic axis by testosterone in adult males: evidence
with growth hormone and cortisol levels in healthy men. for the role of aromatization. J Clin Endocr Metab 76,
JAMA 284, 861–868. 1407–1412.

153

Downloaded from https://www.cambridge.org/core. University of New England, on 17 Feb 2019 at 09:24:42, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/9781108149938.009

You might also like