Download as pdf or txt
Download as pdf or txt
You are on page 1of 50

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/301355622

Neuroendocrine Regulation of Growth Hormone Secretion

Article in Comprehensive Physiology · March 2016

CITATIONS READS
2 1,175

1 author:

Jacques Epelbaum
French National Centre for Scientific Research
441 PUBLICATIONS 20,966 CITATIONS

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Constitutional Thinness View project

Neurobiological roles of somatostatin receptors in normal and pathological aging View project

All content following this page was uploaded by Jacques Epelbaum on 18 April 2016.

The user has requested enhancement of the downloaded file.


Neuroendocrine Regulation of Growth
Hormone Secretion
Frederik J. Steyn,1 Virginie Tolle,2 Chen Chen,3 and Jacques Epelbaum*2

ABSTRACT
This article reviews the main findings that emerged in the intervening years since the previous
volume on hormonal control of growth in the section on the endocrine system of the Handbook
of Physiology concerning the intra- and extrahypothalamic neuronal networks connecting growth
hormone releasing hormone (GHRH) and somatostatin hypophysiotropic neurons and the integra-
tion between regulators of food intake/metabolism and GH release. Among these findings, the
discovery of ghrelin still raises many unanswered questions. One important event was the appli-
cation of deconvolution analysis to the pulsatile patterns of GH secretion in different mammalian
species, including Man, according to gender, hormonal environment and ageing. Concerning
this last phenomenon, a great body of evidence now supports the role of an attenuation of the
GHRH/GH/Insulin-like growth factor-1 (IGF-1) axis in the control of mammalian aging. © 2016
American Physiological Society. Compr Physiol 6:687-735, 2016.

Introduction govern patterned GH release, and thus neuroendocrine control


of GH output.
The aim of the present article is not to recapitulate the
enormous amount of research that followed the discovery of,
the neurohormones that directly regulate GH secretion from
Feedback Control of GH
the pituitary. This has been fully covered in chapters 8 to 12 The secretion of GH from the anterior pituitary gland is regu-
in the volume V on hormonal control of growth in Section 7, lated by complex homeostatic interactions, mediated by neu-
the endocrine system, of the Handbook of Physiology: ral and peripheral influences. These regulators of GH release
Growth hormone-releasing hormone: discovery, regulation, define the amount of GH released during a single secre-
and actions (160), somatostatin (507), growth hormone tory event, and the frequency at which these events occur.
releasing peptides (55), feedback regulation of growth This results in the pulsatile appearance of GH in circulation
hormone secretion (564), and the growth hormone secretory (Fig. 1). The collective interactions between all endogenous
pattern and statural growth (438). Rather, this article will regulators of GH release contribute to interim (ultradian) and
describe main findings that emerged in the intervening enduring (circadian and infradian) fluctuations in the amount
years concerning the intra- and extrahypothalamic neuronal and patterning of GH release. This pattern of GH release is
networks connecting growth hormone releasing hormone conserved across all mammalian species characterized to date,
(GHRH) and somatostatin hypophysiotropic neurons and the although the amount and frequency of secretory events may
integration between regulators of food intake/metabolism vary dramatically between species.
and GH release, among which the discovery of ghrelin still While the clearance of GH partially contributes to the
raises many unanswered questions. One important event was intermittent peaks and troughs in circulating GH, feedback
the application of deconvolution analysis to the pulsatile mechanisms largely modify GH release relative to phys-
patterns of GH secretion in different mammalian species, iological demand. To understand the complex interplay
including man, according to gender, hormonal environment,
and ageing. Application of mathematical models (including * Correspondence to jacques.epelbaum@inserm.fr
deconvolution analysis) to define parameters of GH release 1 University of Queensland Centre for Clinical Research and the
provides a proxy, unbiased by human interpretation, to define School of Biomedical Sciences, University of Queensland, St. Lucia,
key characteristics of GH release. While many observations Brisbane, Queensland, Australia
2 Unité Mixte de Recherche en Santé 894 INSERM, Centre de
focus predominantly on the amount of GH release, patterning
Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne
of pulsatility largely defines the physiological actions of GH, Paris Cité, Paris, France
contributing in the tissue-specific divergence in GH action. 3 School of Biomedical Sciences, University of Queensland, St. Lucia,
This patterning is largely controlled via the hypothalamus, Brisbane, Queensland, Australia
reflecting interactions between GH pulse generators. Proxy Published online, April 2016 (comprehensivephysiology.com)
measures of pulsatility (through deconvolution) are critically DOI: 10.1002/cphy.c150002
important should we hope to define neuronal interactions that Copyright © American Physiological Society.

Volume 6, April 2016 687


Growth Hormone Axis Comprehensive Physiology

Ultradian

Infradian
Home Long-loop feedback
ostatic
Ultrashort loop Pressur
Short-loop feedback es

Peak
Peak

Trough
Trough

Trough

Hours Days/weeks/months Years Time

Figure 1 Contribution of feedback regulation to GH release. Ultrashort and short-loop feedback mech-
anisms contribute largely to the generation of a single GH secretory event, modulating output over hours.
This culminates in the ultradian pulsatile appearance of GH in circulation, defined by peaks and troughs
in GH release. In turn, long-loop feedback mechanisms contribute to the infradian regulation of secretion,
modulating peak GH release and patterning over extended periods of time. Given homeostatic pressures
(metabolic, reproductive, age associated), cross-talk between short and long-loop mechanisms (illustrated
as white double arrows) ensure physiological GH secretion. For this, long-loop feedback mechanisms will
modulate ultradian responses, whereas ultradian patterns may impact infradian feedback.

between factors that regulate the release of GH, one must first long-loop feedback); however, it must be noted that IGF-1 is
define the nature of the various feedback mechanisms that not the sole regulator of long-lasting changes in GH release.
contribute to the regulation of GH release in general. These Many factors that contribute to infradian GH secretion are
interactions are varied and, given the complexities associated discussed throughout the remainder of this review. For a fur-
with the release of GH, it is not feasible to provide a detailed ther comprehensive overview of feedback mechanisms, please
overview of all interactions that shape GH release. Rather, we consult prior reviews on this topic (174, 546), or Chapter 12
will provide an overview of the most commonly associated in the volume on hormonal control of growth in the section on
feedback mechanisms thought to contribute to ultradian and the endocrine system of the Handbook of Physiology (438).
infradian patterns of GH release. To account for life-long The nature an interaction sites of feedback interactions spe-
changes in GH release, we will first provide an overview of cific to the GH axis are summarized in Figure 2. As illustrated
mechanisms that shape ultradian GH secretory events before in this article, our understanding of GH feedback and mech-
extending the discussion to factors that shape infradian GH anisms that define physiologically relevant GH output are
release. While ultra short-loop feedback and short-loop feed- continuously evolving.
back mechanisms generally shape ultradian, and long-loop
feedback mechanisms generally shape infradian GH secretion
profiles, true homeostatic control of GH release is defined by Ultrashort-loop feedback control of GH release
the collective interactions of these mechanisms. In this way, During ultra short-loop feedback, secretory products from
long-loop feedback pressure can define the nature of ultra a cell feed back to the same cell to modify the function of
short- and short-loop feedback (and thus daily GH secretion the cell itself. This is often referred to as “autoregulation”
patterns), whereas ultrashort- and short-loop feedback or “autocrine regulation.” In this instance, pituitary soma-
ultimately impact infradian GH secretion, and thus long-loop totrophs respond to GH, whereas somatostatin and GHRH
feedback control of GH release. This is schematically neurons may respond to somatostatin and GHRH, respec-
illustrated in Figure 1. (174, 546) tively. Ultrashort-loop feedback shapes somatotroph activity,
Somatostatin and GHRH are predominant factors that and somatostatin and GHRH neuronal activity, contributing
regulate pulsatile GH release, and thus this section will to ultradian patterns of GH release. In addition, long-acting
predominantly detail interactions between somatotrophs and effects of ultrashort-loop feedback mechanisms may con-
hypothalamic somatostatin and GHRH neurons (as examples tribute to infradian variations in GH release, or a derangement
of ultrashort-loop and short-loop feedback). We extend obser- in GH secretion patterning. For example, overexpression of
vations to feedback effects from IGF-1 (as an example of GH receptors (GH-R) in mice contributes to somatotroph

688 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

pendent of somatostatin. Presumably, this was in response


to exogenous GHRH-mediated suppression of GHRH neu-
ronal activity, reflecting the removal of GH stimulation. While
these observations were corroborated (311), demonstrating a
marked decrease in circulating levels of GH following cen-
tral administration of low dosages of GHRH (2 and 20 ng),
interpretation of the initial suppression of circulating GH fol-
lowing GHRH (505) may have been compromised by the pres-
ence of high levels of corticotrophin-releasing-factor (CRF)
in the “GRF” preparation (516). As discussed later on in
this article, CRF is a potent inhibitor of GH release, acting
peripherally and centrally (436). In light of this, it appears
that initial observations that documented the central actions
of GHRH on GH release may not involve autocrine feed-
back of GHRH to inhibit pituitary GH release. Indeed, injec-
tion of higher dosages of GHRH into the third ventricle
increase circulating levels of GH, suggesting that the per-
ceived potential positive autocrine effects of GHRH may in
fact have occurred as a consequence of exogenous GHRH
Figure 2 Feedback mechanisms. The release of GH from the ante- crossing the median eminence and directly interacting with
rior pituitary gland is mediated through feedback mechanisms, act- pituitary somatotrophs (311). Given historical difficulties to
ing on three levels. Somatostatin and GH release is controlled through define the autocrine actions of GHRH within the hypotha-
ultrashort-loop feedback (orange arrows), whereby somatostatin and
GH modulates it own release. While ultrashort-loop feedback is pro- lamus, potential in vivo GHRH autoregulation requires fur-
posed for GHRH, evidence for this is not conclusive (illustrated by ther validation. Moreover, current measures cannot account
broken line arrow). GH pulsatility is regulated by short-loop feedback for possible indirect somatostatin-mediated effects, and thus
between somatostatin, GHRH and GH (blue arrows). GH acts within
the hypothalamus to stimulate somatostatin and possibly inhibit GHRH alterations in GHRH function as a consequence of GHRH-
release in the median eminence (ME; note, for GHRH neurons, com- mediated somatostatin feedback (see somatostatin-mediated
pelling evidence for GH feedback does not exist). Once released into short-loop feedback). Surprisingly, the development and char-
portal circulation, somatostatin travels to the pituitary gland where it
inhibits GH release, whereas GHRH stimulates GH release. In addition, acterization of transgenic mice that express green fluorescent
somatostatin inhibits GHRH activity, and it is thought that GHRH neu- protein (GFP) in GHRH neurons (23) has not yet heralded
rons may stimulate somatostatin neuronal activity, and therefore somato- studies that address autocrine regulation of GHRH activity.
statin release. The release of GH relative to physiological demand
is entrained through a number of peripheral factors. In this instance, Somatostatin. Compared to GHRH, potential autocrine
long-loop feedback (illustrated in green arrows) from insulin-like growth effects between somatostatin and somatostatin producing neu-
factor-1 (IGF-1, produced in the liver in response to GH) results in rons are better established. Somatostatin analogues modify
the suppression of GH release through the stimulation of somatostatin
release, and the inhibition of GH and GHRH release. somatostatin release from isolated hypothalamic cell cultures,
suppressing somatostatin release (411). Up to 55% of somato-
statin expressing neurons in mouse hypothalamic neurons in
atrophy and hypoplasia (480, 481), and presumably a persis- culture express sst1 receptors while sst2 receptor expression
tent derangement in GH secretion pattern. To account for the amounts to roughly 40% (422) and the inhibitory autofeed-
collective impact of short- and long-loop feedback effects back of somatostatin is likely to depend on sstr1 receptors
on the neuroendocrine control of GH release, one must first (287). Very low dosages of icv-administered somatostatin
define the nature of autocrine regulation within somatostatin stimulate GH release (577). Accordingly, somatostatin may
and GHRH neurons and somatotrophs. promote the release of GH by suppressing its own release.
While directly promoting the hypersecretion of GH, these
effects cannot account for somatostatin-induced suppression
Hypothalamic ultrashort-loop feedback: GHRH
of GHRH (see somatostatin and GHRH mediated short-loop
and Somatostatin
feedback).
GHRH. Initial observations showed a marked inhibition of
circulating GH following icv treatment with human pancre-
Pituitary ultrashort-loop feedback: Growth Hormone
atic growth hormone-releasing factor (504) (To be consistent,
GHRH is used in this article as the sole acronym though Somatotrophs express GH receptors (152), suggesting a
the neurohormone was initially known as growth-hormone- functional role for GH in mediating somatotroph-specific
releasing factor (GRF, GHRF), and later as somatoliberin GH release. Potential autocrine and paracrine effects of
or somatocrinin]. This suppressive effect persisted regardless IGF-1 within the pituitary gland confound functional studies
of pretreatment with somatostatin antiserum, suggesting that that define the autocrine effects of GH on somatotrophs.
central administration of GRF inhibited GH secretion inde- Nonetheless, compelling evidence from isolated primary

Volume 6, April 2016 689


Growth Hormone Axis Comprehensive Physiology

somatotrophs (175, 444) and GH secreting cell-lines (includ- Short-loop feedback control of GH release
ing GH3 cells) (289, 444) demonstrate direct inhibitory
Short-loop feedback is defined by regulation of hypotha-
actions of GH on somatotrophs. In all assessed cases,
lamic factors by pituitary hormones or other hypothalamic
GH modulates basal GH release without altering GHRH
hormones. For the GH axis this involves the feedback of
mediated GH secretion. These observations highlight a very
GH to mediate hypothalamic regulators or interactions
critical caveat when using cell lines and isolated primary
between hypothalamic regulators (paracrine regulation), in
cells to model somatotroph interactions (289). Inevitably,
this instance GHRH and somatostatin. Hallmark studies
the accumulation of GH in culture will inhibit GH release,
by Brazeau and Vale (60) in Guillemin’s laboratory and
thereby altering cell responses during prolonged incubation
Tannenbaum and Ling (513) first demonstrated interactions
periods or when changing incubation conditions. These
between somatostatin and GHRH. It was observed that GHRH
concerns are further highlighted by numerous in vivo studies
failed to induce the release of GH in rats when somatostatin
confirming that GH can regulate its own transcription,
levels were thought to be elevated (this was confirmed by pre-
translation and secretion (564). Given the complexities of
treatment with antisomatostatin antibodies) (513). From these
the GH axis, and the existence of numerous interacting
studies, it was proposed that somatostatin and GHRH are
feedback mechanisms, the exact direct effect of GH feedback
released alternately. This was confirmed (415), and provided
on somatotroph specific GH release is hard to define. This is
the premise for many of the studies documenting short-loop
further highlighted by diverging observations between in vitro
interactions between these key hypothalamic regulators
and in vivo measures. For example, GH does not modify
of pulsatile GH release.
GHRH mediated GH release from isolated somatotrophs
(175, 444), whereas exogenous GH blunts the pituitary GH
response to GHRH (417). In vivo, the observed actions of
GH on GH release may occur in response to direct actions of
Hypothalamic short-loop feedback: GHRH and
Somatostatin
GH on somatotrophs, or more likely, through indirect actions
including IGF-1 or somatostatin mediated effects. GHRH and somatostatin are thought to act in concert to reg-
Recent advances in in vivo imaging methodologies con- ulate the production and release of GH. As part of this feed-
firmed that structural and not only pharmacological inter- back interaction, GHRH and somatostatin may also modulate
actions between somatotrophs might facilitate the autocrine the synthesis and secretion of each other through bidirec-
regulation of GH release. In the mouse, interactions between tional hypothalamic interactions. Initial observations mapped
somatotrophs are established at a very young age (embryonic the distribution of hypophysiotropic somatostatin producing
day 15.5), with clusters of somatotrophs forming a continuum neurons in the anterior periventricular nucleus (PevN) of
of cells during the growth of the pituitary gland (238). These the hypothalamus (see also 507). Moreover, somatostatin-
clusters are interconnected by strands of somatotrophs (53), producing neurons are also located within the dorsomedial
joined through highly specialized gap junctions that medi- portion of the ARC, with axons projecting to the ventrolat-
ate calcium-signaling events that facilitate network activity. eral portion of the nucleus (the location of GHRH neurons).
Indeed, it is thought that this arrangement may provide a sub- Dual labeling revealed the juxtaposition of these somato-
stratum for long-term or sustained changes in somatotroph statin fibers with dendrites and cell bodies of GHRH neu-
network activity (294). While a comprehensive assessment of rons (304). This was corroborated by subsequent studies
in vivo GH output relative to somatotroph clustering is needed, (219, 576). Morphological assessment of these interactions
substantial changes in GH output in mice occur during peri- revealed synaptic interactions with GHRH neurons (219,304),
ods of natural reorganization of the somatotroph network. For suggesting that the physiological interactions that mediate the
example, the reorganization of the somatotroph network is pulsatile release of GH originate within the hypothalamus.
sustained following birth, and extensive clustering of soma- Of interest, combined retrograde tracing of somatostatin and
totrophs in male mice occurs between 40 and 70 days of GHRH neurons revealed a very limited number of co-labeled
age (452). This period of development is characterized by somatostatin fibers following Fluorogold injections into the
the establishment of pulsatile GH secretory patterns remi- basal hypothalamus, whereas occasional GHRH fibers were
nisce of adult male mice (503). Therefore, it is thought that doubly labeled following Fluorogold injections to the pre-
pubertal maturation of GH release, while mediated through optic anterior hypothalamic area (576). Indeed, subsequent
hypothalamic regulators, occurs in response to reorganization observations questioned interactions between somatostatin
of autocrine somatotroph interactions (293). This process is and GHRH neurons, proposing the absence of major direct
facilitated by feedback from gonadal hormones, as illustrated connections between hypophysiotropic GHRH and somato-
by the loss of organization following prepubertal castration statin neurons (146). While questioning interactions between
(53). For a comprehensive overview of network arrangements GHRH and somatostatin neurons, these observations further
within the pituitary gland, the relationships between soma- suggest that GHRH may not exert the same direct level of
totrophs, blood vessels and other endocrine cells, and potential control on somatostatin neurons as somatostatin neurons do
mechanisms of information exchange between somatotrophs, on GHRH neurons. Indeed, other neuropeptides, including
please refer to (294). neuropeptide Y and POMC derived ß-endorphin and α-MSH

690 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

(145) may indirectly facilitate GHRH-induced stimulation of region of the ventromedial nucleus and the paraventricu-
somatostatin. lar nucleus (38, 70, 348). Moreover, hypophysiotropic PevN
In line with the respective inhibitory and stimulatory somatostatin neurons express GH receptors (68, 349). This
roles of somatostatin and GHRH on GH release, one may provides a structural premise for the potential regulation of
anticipate that somatostatin neurons inhibit GHRH activity GHRH and somatostatin release by GH. While the mecha-
whereas GHRH may stimulate somatostatin neurons. Indeed, nisms that accounts for transport of GH into the brain remains
removal of somatostatin increases GHRH message in the rat unclear, peripheral GH does cross the blood brain barrier (399)
(42), and treatment of perifused hypothalamic tissue with supporting potential direct regulatory effects.
somatostatin inhibits basal GHRH release (591). Conversely, While it is thought that feedback between GH and its prin-
exposure to GHRH promotes somatostatin content and release cipal hypothalamic regulators may largely contribute to the
from cultured primary hypothalamic cells (118). In an effort autoregulation of GH release, functional measures of activa-
to better define interactions between somatostatin and GHRH tion of somatostatin and GHRH neurons following GH admin-
neurons, Katakami and colleagues showed that icv treatment istration suggest otherwise. Indeed, GH may only impact a
with hGHRH suppresses anticipated spontaneous GH pulses small subset of somatostatin and GHRH expressing neurons.
(251). This suppressive effect of GHRH was prevented For example, rGH injections in the rat contributes to less than
following pretreatment with antisomatostatin antiserum, 11% of activation of GHRH neurons and only 5% activa-
indicating the presence of an inhibitory feedback system that tion of somatostatin neurons as measured by cFos expression
governs GHRH-mediated GH release. Subsequent observa- (69). As detailed later, this limited interaction may be suffi-
tions confirmed that icv treatment with GHRH induced a cient to modulate the output of GH relative to physiological
significant rise in portal plasma concentrations of somato- demand; however, it is important to note that other regula-
statin and a rise in somatostatin production rate (351). In line tors of GH release may facilitate hypothalamic GH feedback.
with this, isolation of somatostatin neurons from feedback For example, the majority of arcuate neuropeptide-Y (NPY)
from GHRH neurons [through lesion of the arcuate nucleus expressing neurons in the rat coexpress the GH receptors
(ArcN)] resulted in an overall decrease in somatostatin tone, (82,349), while icv administration of NPY inhibit GH release
and a decrease in somatostatin content within the median (429), presumably through promoting somatostatin release
eminence (454). This was further corroborated by a decrease (349). Short-loop feedback interactions specific to hypotha-
in somatostatin content in the median eminence following lamic somatostatin and GHRH release that account for the
immunoneutralization of GHRH neurons with anti-GHRH pulsatile release of GH from the anterior pituitary gland are
antibodies (118). While providing convincing evidence to summarized in Figure 3.
demonstrate a clear relationship between somatostatin and Elevated GH concentrations are associated with a decrease
GHRH in mediating GH release, the true physiological in hypothalamic GHRH mRNA expression (43, 91, 116, 117);
interactions between these factors need careful validation. however, this may not contribute to altered GHRH release.
For example, within sheep withdrawal of somatostatin does For example, an elevation in endogenous GH tone is asso-
not necessarily precede GH pulses (75,525), and somatostatin ciated with a decrease (39, 318) or no change (245) in
release may rise or remain unchanged immediately prior to hypothalamic GHRH content, while GH treatment increases
a rise in circulating levels of GH (75). Derangements in the hypothalamic GHRH content (296, 343). GH deficiency is
proposed relationship between somatostatin, GHRH and GH associated with a reduction in hypothalamic GHRH content
may reflect the complexity of interplay between other key (164, 253, 337), and this reduction in GHRH content is par-
regulators. Similar arguments against the existence of a sim- tially reversed following GH treatment (164,253). It should be
plified feedback interaction between these factors are seen in noted, that while an increase in GHRH content and decrease in
other species, including the rat (296). For the purpose of this GHRH mRNA is consistent with an inhibition of GH release,
section, the majority of factors that modify GH release, and hypothalamic GHRH content likely reflect the net difference
neuronal populations that contribute to the central regulation between GHRH synthesis and release. Therefore, observa-
of GH release were largely ignored. This allows an opportu- tions that contrast hypothalamic GH content with peripheral
nity to describe feedback interactions between somatostatin, GH release, but that do not provide measures of synthesis
GHRH and GH; factors deemed as principal regulators of and release of GHRH must be cautiously interpreted. More-
GH release. Other regulators of GH release are discussed over, the effects of GH on GHRH mRNA or content vary
throughout the remainder of this article and can also be found relative to age and gender. In aged rats (20 months old),
in chapter 11 from Handbook of Physiology Section 7, the treatment with GH does not suppress hypothalamic GHRH
endocrine system, vol V Hormonal control of growth (564). mRNA expression (117), whereas the negative effects of GH
on GHRH content in female rats is greatly diminished (318).
The lack of consistent changes in altered GHRH output rela-
Pituitary short-loop feedback: Growth Hormone
tive to GH suggests that feedback between GH and somato-
Growth hormone receptors are distributed throughout the statin, and consequential changes in somatostatin release may
hypothalamus, with mRNA expression detected within the more likely underpin hypothalamic events (including GHRH
arcuate nucleus, PevN, supraoptic nucleus, the ventrolateral content and release) that regulate episodic GH release, and

Volume 6, April 2016 691


Growth Hormone Axis Comprehensive Physiology

Figure 3 Interactions between somatostatin and GHRH that account for the pulsatile pattern of GH secretion. Comprehensive in vivo and in vitro
assessment of the production and release of GH has resulted in the evolution of theory of peripheral and centrally mediated mechanisms that
modulate the pulsatile release of GH. Periods of prevailing low levels of circulating GH (1) are associated with reduced somatostatin release into
pituitary portal vasculature (2). The withdrawal of somatostatin inhibition of GHRH neurons (3) and somatotrophs (3) culminates in the release
of GHRH into pituitary portal vasculature (4), and the stimulation of GH release (5). Periods of prevailing elevated levels of circulating GH (6)
are associated with the activation of somatostatin-expressing neurons (7) and inactivation of GHRH-expressing neurons (7). The resulting rise in
somatostatin activity contributes to the direct inhibition of GHRH-neuronal activity (8) and the release of somatostatin into portal vasculature (8), cul-
minating in reduced somatotroph activity (9) and the cessation of the release of GH into peripheral circulation (10). Complex interactions between
central, pituitary and peripheral factors may disrupt these hypothalamic interactions, resulting in physiologically relevant GH release. Colored
arrows indicate selective activation of mechanisms coupled with prevailing low/basal (left schematic) or elevated/peak (right schematic) levels
of peripheral circulating GH. Green arrows indicate stimulatory pathways. Orange arrows indicate inhibitory pathways. Additional abbrevia-
tions: GHR, growth hormone receptor; SRIF-R, somatostatin receptor; GHRH-R, growth hormone releasing hormone receptor; PevN, periventricular
nucleus; ArcN, arcuate nucleus; PG, pituitary gland.

may be dependent on other factors that modulate GH release extent, a reduction in endogenous circulating GH levels result
relative to age and reproductive function. in an increase in hypothalamic somatostatin content (39,246),
As with GHRH, reported effects of GH in modulating whereas exogenous GH treatment results in an increase in
hypothalamic somatostatin mRNA and protein content and hypothalamic somatostatin content (343). While a persistent
release varies considerably. This is underpinned by the decrease in circulating GH levels contributes to a reduction
potential for other factors to modulate somatostatin activity, in hypothalamic somatostatin mRNA expression (43),
a shift in the timing between GH signaling and somatostatin somatostatin mRNA expression may not accurately reflect
transcription and release, and the heterogeneous nature of altered somatostatin or GH release at that time. For example,
somatostatin assessment. Nonetheless, the bulk of observa- the immediate cessation of pulsatile GH release in mice
tions suggest that GH stimulates somatostatin release. To this following food withdrawal is associated with an initial rise

692 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

in somatostatin mRNA expression within the arcuate/PevN to single one off samples (162) or extrapolation of pulsatility
complex, whereas somatostatin mRNA expression is greatly by rank-plot analysis (163). Regardless, these observations
diminished following an extended period of suppressed agree with initial observations demonstrating a direct rela-
GH release (487). This biphasic response in somatostatin tionship between the abundance of IGF-1R on somatotrophs
mRNA expression likely reflects the role of somatostatin in and GH release (587), and the attenuation of GH release in
suppressing GH release in fasting rodents (509), whereas the response to IGF-1 (587).
subsequent reduction in somatostatin mRNA content occurs While the actions of IGF-1 within the pituitary gland are
in response to diminished feedback from GH needed to sus- well defined, the role of IGF-1 in regulating both hypotha-
tain somatostatin activity (as discussed earlier). The in vitro lamic somatostatin and GHRH action remains somewhat
assessment of hypothalamic somatostatin release in response unresolved. The IGF-1 receptor is abundantly expressed
to GH treatment confirms this feedback relationship, wherein throughout the hypothalamus, including the ArcN, dorsal and
GH exposure stimulates hypothalamic somatostatin content ventromedial nuclei of the hypothalamus, and the median
and release (5, 39, 117, 206, 353, 472). Indeed, it is thought eminence (573). It thus seems likely that IGF-1 will inter-
that somatostatin is the primary feedback mechanisms for the act on different levels to modulate GH release. Intracere-
rhythmic release of GH in rodents (558). For this notion to broventricular administration of IGF-1 in rats results in a
be correct, one must however assume that rhythmical activity decrease in GH pulse amplitude (1, 511), an increase in
from GHRH neurons exist independent of feedback from GH hypothalamic somatostatin mRNA expression, and a decrease
(558), a premise not entirely confirmed as yet. in hypothalamic GHRH mRNA expression (453). By contrast,
peripheral injections of IGF-1 in the GH deficient dwarf rat
do not impact hypothalamic somatostatin or GHRH mRNA
Long-loop feedback control of GH release expression (453). This suggests divergent and independent
A large number of peripheral factors impact GH secretion, actions of IGF-1 on hypothalamic and pituitary mediated
ensuring the entrainment of GH release relative to physio- GH release. Periodic GH pulse frequency is largely depen-
logical demand. While contributing to the patterned release dent on GHRH release (159, 237, 415), and thus the observed
of GH, these factors generally modify the production and reduction in GH pulse frequency following sustained IGF-1
release of GH over weeks, months, and years. In this regard, infusion in humans is potentially due to an attenuation of
long-loop feedback within the GH axis is defined by the reg- GHRH secretion. This is in line with the suppression of
ulation of other hormones or molecules that are secreted from GHRH synthesis and secretion in rat hypothalamic tissue fol-
extrahypothalamic or pituitary sites. For the purpose of this lowing IGF-1 treatment (473), and in rats in vivo following
section, we will only discuss the role of IGF-1 in regulat- icv IGF-1 treatment (453). Given the magnitude of the sup-
ing GH release, however numerous other factors participate pressive effects of IGF-1 infusion on pulsatile GH release
in this process. These are discussed in detail throughout the in humans, it is unlikely that this diminished release of GH
remainder of this article. It is important to note that long- occurs solely in response to altered GHRH feedback. Indeed,
loop regulators of GH release may not interact with all pri- initial observations demonstrated a stimulatory role for IGF-1
mary components of the hypothalamic/GH axis, and therefore (termed somatomedin-C at the time) on hypothalamic somato-
may solely impact somatostatin, GHRH, and/or somatotroph statin release, wherein purified somatomedin-C (218) pro-
activity. Regardless, the actions of IGF-1 on GH release are moted the dose-dependent release of somatostatin from intact
inhibitory, and generally reflected by observed inhibitory rat hypothalamic tissue (40). Subsequent observations con-
effects of IGF-1 on somatotrophs and GHRH neurons, firmed that continuous infusion of IGF-1 in fed and fasting
and stimulatory effects of IGF-1 on somatostatin neurons males inhibited pulsatile GH release, primarily through the
(Fig. 2). reduction of spontaneous GH pulse amplitude and pulse fre-
IGF-1 is expressed within the pituitary gland (137). Thus, quency (41). Moreover, IGF-1 infusion attenuated GHRH-
while more commonly considered an endocrine regulator of induced GH release, suggesting that IGF-1 may promote
GH release, IGF-1 may act in a paracrine fashion to regulate somatostatin release to inhibit GH secretion. While describing
GH release. The role of IGF-1 as a paracrine regulator of GH a simple feedback mechanism whereby IGF-1 modulates GH
release will not be discussed here. Rather, this section will release, these observations highlighted the complex nature
focus on established interactions between liver-derived IGF- through which IGF-1 is thought to modulate hypothalamic
1 and primary components of the GH axis; somatotrophs and and pituitary control of GH release (Fig. 2).
somatostatin or GHRH releasing neurons.
IGF-1 treatment inhibits GH gene transcription, GH Central Regulation of GH Secretion
mRNA expression and GH release from isolated somatotrophs
(357, 590). In vivo, selective deletion of IGF-1 receptor (IGF- Many neurotransmitters and neuropeptides modulate the
1R) expression on somatotrophs results in an overall rise in activity of the GH/IGF-1 axis by interacting with GHRH and
circulating levels of GH (162). The nature of this change in GH somatostatin or by directly acting at the level of the soma-
pulsatility remains undefined, as current measures are limited totroph to modify GH release. This has been extensively

Volume 6, April 2016 693


Growth Hormone Axis Comprehensive Physiology

reviewed elsewhere (174, 326, 536) and only the most vali- L-DOPA (L-3,4-dihydroxyphenylalanine). In primates,
dated ones are described herein. catecholamines stimulate GH secretion. Indeed, oral admin-
istration of L-DOPA, the precursor of both NE and DA,
causes release of GH in humans (56). Evidence for nora-
Neuromodulators drenergic control is derived from experiments showing that
Monoamines [Dopamine (DA), Norepinephrine (NE), Sero- L-DOPA-induced GH release is blocked by phentolamine, an
tonin] are of considerable importance in the neural reg- α-adrenergic blocking agent (247). In addition, clonidine, a
ulation of GH secretion. The high concentration of these central α-agonist, promotes GH release in men (284). GH is
monoamines in the hypothalamus, particularly in the median also released in men by subemetic doses of apomorphine, a
eminence, suggests that monoamines have important regula- centrally active dopaminergic stimulator (64). This does not
tory functions for the release of the hypothalamic hypophys- seem to be a stress response, as it is not accompanied by a rise
iotropic hormones. Here we provide a general overview of in plasma cortisol (64, 283). Both L-DOPA-induced (347)
the role of monoamines in the regulation of GH output, and apomorphine-induced (136) GH release are attenuated
focusing selectively on catecholamines, serotonin, and acetyl- by prior glucose administration, which indicates that glucore-
choline (Fig. 4). For a comprehensive overview of the role ceptor stimulation can partially override catecholaminergic
of monoamines in regulating GH output, please refer to stimuli for GH release.
(133, 174, 327, 362, 536). Dopamine. The specific involvement of dopamine in the
regulation of GH secretion is not clearly demonstrated. Its
Catecholamines action seems to be mediated in the pituitary gland rather than
in the hypothalamus. Indeed, dopamine or a D1-R agonist
The role of catecholamines in the regulation of GH secretion induces a transitory increase in GH release both in vitro from
has been extensively investigated since the observation that pituitary cells in culture and in vivo after destruction of the
GH secretion was inhibited in animals treated with an inhibitor mediobasal hypothalamus (49). In contrast, dopamine would
of tyrosine hydroxylase biosynthesis (130). rather exert an inhibitory effect in the hypothalamus since
it stimulates somatostatin release by hypothalamic explants
(49).
Adrenaline and noradrenaline. Both adrenaline and nora-
drenaline play a major role in the regulation of GH secretion
under the stimulatory control of α2-adrenergic receptors
and inhibitory control of α1- and β-adrenergic receptors.
α2-Adrenergic agonists, such as clonidine, stimulate GH
secretion in both humans (49) and rats (49, 130, 446) and
α2-adrenergic antagonist, yohimbine, blocks the increase
in GH induced by clonidine. In contrast, methoxamine, an
α1-adrenergic agonist, reduces the amplitude of GH secretory
peaks in the rat (47) and isoproterenol, a β-adrenergic agonist,
inhibits GHRH-induced GH secretion (279).
Evidence from intravenous administrations of pharma-
cological agents to urethane-anesthetized rats suggests that
β-receptors are important in the regulation of GH release
(255). Intravenous administration of chlorpromazine elic-
its GH release, an effect that is blocked by propranolol.
The chlorpromazine response is attenuated by hypothalamic
lesions and by posterior hypothalamic cuts, suggesting that
Figure 4 Hypothalamic control—neuromodulators. The control of
neural inputs to the hypothalamus are important for the medi-
GH release from somatostatin and GHRH-expressing neurons is medi-
ated through interactions with hypothalamic neuromodulators. In this ation of monoaminergic GH response in rodent species (254).
instance, dopamine (DA) and adrenaline [acting through selective There is little evidence suggesting that monoamines reg-
activation of the α1-receptor subtype (α1-R)] stimulates somatostatin- ulate GH secretion directly at the pituitary level. Rather,
neuronal activity, thereby inhibiting GH release. Adrenaline [acting
through selective activation of the α2-receptor subtype (α2-R)], serotonin monoamines may function as neurotransmitters in the relay
(5-HT), and acetylcholine (Ach) stimulate GH release, through inhibi- of these responses to GHRH and somatostatin neurons. α2-
tion of somatostatin-expressing neurons. By comparison, activation of adrenergic receptor agonists may act via stimulation of GHRH
GHRH-expressing neuronal activity by adrenaline (acting through selec-
tive activation of the α2-R) and 5-HT results in increased GH output. In release. Indeed, clonidine stimulates GHRH release from per-
turn, acting through select activation of the β2-receptor subtype (β2-R), ifused rat hypothalamic explants (241) and is not able to
adrenaline inhibits GH release by inhibiting GHRH-neuronal activity. stimulate GH secretion in rats treated with an anti-GHRH
Stimulatory interactions are highlighted in green arrows. Inhibitory inter-
actions are highlighted in orange arrows. Additional abbreviations: antibody (344). In addition, clonidine would act by inhibiting
ME, median eminence. somatostatin since it potentiates GHRH-induced GH response

694 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

in rats in conditions of elevated somatostatin tone (299, 300) Neuropeptides


as well as in humans (127). α1/β-adrenergic receptor ago-
Neuropeptide Y
nists would regulate GH secretion by modulating somatostatin
release. Immunoneutralization of somatostatin in young rats Neuropeptide-Y is a 36 amino-acid peptide (520) that belongs
completely prevents GH secretion induced by methoxamine, to the family of pancreatic polypeptides. Hypothalamic neu-
an α1-adrenergic receptor agonist. In addition, in both rats rons expressing NPY are mostly present within the arcuate,
and humans, GH response to GHRH is potentiated by pro- paraventricular, and the periventricular nuclei (113) (Fig. 5).
pranolol, a β-adrenergic receptor antagonist (258) that can GH release is considerably diminished following central NPY
inhibit somatostatin release in vitro (433). In contrast, a β2- treatment (76, 192), with chronic administration of NPY into
adrenergic receptor agonist, isoproterenol, suppresses GH the lateral ventricle contributing to a reduction in circulating
response to GHRH, and its GH-inhibiting effect is blocked levels of GH (413) and IGF-1 (425). Central administration of
by an antisomatostatin antibody (279). These data demon- NPY also silences the pulsatile release of GH. This effect is
strate that inhibition of GH secretion induced by α1- and
β-adrenergic receptors are partly mediated by an increase in
the somatostatin endogenous tone (433).

Serotonin
In the rat, icv administration of serotonin increases plasma GH
concentrations (556) whereas inhibitors of serotonin synthesis
or serotonin receptors antagonists reduce plasma GH concen-
trations. Serotonin has no effect in the pituitary gland (256)
but modulation of hypothalamic somatostatin and GHRH
release is suggested by the presence of serotoninergic fibers
in PevN and ArcN nuclei. Indeed, serotonin inhibits somato-
statin release in vitro from perifused rat hypothalamic explants
(432) and in vivo, the serotonin-induced GH increase is sup-
pressed by an anti-GHRH antibody (365).
Oral administration of L-Tryptophan or 5-
hydroxytryptophan (5-HTP) in men causes a mild GH
release (229). In addition, the GH response to hypoglycemia
is blocked by the serotonin antagonists methysergide and
cyproheptadine (46, 475). Administration of 5-HTP is also
effective in causing GH release in the Rhesus monkey. The
effect of serotonin is observed in all species tested, indicating
a consistent interspecies effect.
Figure 5 Intrahypothalamic interactions—neuropeptides. Complex
intrahypothalamic interactions control the activity of somatostatin-
Acetylcholine expressing and GHRH-expressing neurons, thereby regulating the
pulsatile release of GH. Corticotrophin releasing hormone (CRH)-
Acetylcholine plays an important role in the central control expressing neurons within the paraventricular nucleus (PvN) stimu-
of GH secretion in humans (73) as well as in animals (65). late somatostatin activity, thereby inhibiting GH release. This is mod-
Muscarinic cholinergic agonists stimulate basal GH secre- ulated through activation of CRH-expressing neurons by neuropeptide-
Y (NPY)-expressing neurons within the arcuate nucleus (ArcN), acting
tion and potentiate GH response to GHRH. Conversely, mus- through the Y-1 (Y1-R) and Y-2 (Y2-R) receptor subtypes. CRH activity
carinic antagonists inhibit spontaneous GH secretion and GH is further modulated through activation of the melanocortin receptor
response to GHRH (308, 327). A direct action on the pitu- subtype-4 (MC4-R). NPY-expressing neurons further inhibit GH release
through activation of somatostatin-expressing neurons. Of interest, NPY-
itary is not likely (308) while, at the hypothalamic level, expressing neurons may indirectly promote the release of GH through
acetylcholine acts by blocking hypothalamic somatostatin the activation of pro-opiomelanocortin (POMC)-expressing neurons,
release rather than by stimulating GHRH release. Indeed, which inhibits somatostatin-expressing neuronal activity. This control
of GH release is further modulated through the stimulation of NPY-
somatostatin release from hypothalamus in culture is inhib- expressing neurons by somatostatin (within the ArcN), and through
ited by acetylcholine or neostigmine, a cholinergic agonist activation of GHRH expressing neurons by low levels of somatostatin.
(431). In addition, in animals depleted from somatostatin Stimulatory interactions are highlighted in green arrows. Inhibitory
interactions are highlighted in orange arrows. Additional abbrevia-
by cysteamine or an antisomatostatin antibody, acetylcholine tions: ME, median eminence; SST1, somatostatin receptor subtype-1,
has no effect (308). In contrast, immunoneutralization of SST2, somatostatin receptor subtype-2; SST3, somatostatin receptor
GHRH by a GHRH-specific antibody does not prevent GH subtype-3; SST5, somatostatin receptor subtype-5; Y1-R, Neuropeptide-
Y specific receptor subtype-1; Y2-R, neuropeptide-Y specific receptor
release induced by pyridostigmine, an inhibitor of acetyl- subtype-2; GAL-R1, galanin receptor subtype-1; GAL-R2, galanin recep-
choline esterase (566). tor subtype-2; MC3R, melanocortin receptor subtype-3.

Volume 6, April 2016 695


Growth Hormone Axis Comprehensive Physiology

abolished by anterolateral deafferentation of the medial basal tends to increase (i.e., humans and sheep) or decrease the pul-
hypothalamus, a procedure that disrupts the somatostatiner- satile release of GH (i.e., rats and mice), in spite of apparently
gic input to the MBH (349), suggesting that NPY selectively similar neuroanatomical wiring, the nature of the interaction
modulates hypothalamic regulators that prime the periodic between NPY and hypothalamic regulators of GH release
release of GH through interactions with hypophysiotropic to account for differential effects on GH output remains
PevN somatostatin neurons. Of interest, central injection of completely unexplored.
NPY to specifically target neuronal populations within the The actions of NPY are mediated via a family of high
ArcN and PevN inhibit GH release, whereas similar injec- affinity receptors (407), including Y1R, Y2R, Y4R, Y5R, and
tions of IGF-1 do not alter GH output (349). To this extent, in mice and rabbits, the Y6R receptor (142, 199). It is gener-
NPY activation may trump IGF-1 as a key regulator of cen- ally assumed that the postsynaptic Y1R regulates food intake
trally mediated GH feedback. Indeed, the vast majority (95%) (257), whereas the Y2R inhibits the synthesis and release
of NPY neurons within the ArcN express GH receptors (244), of NPY (62). Central administration of an Y1R agonist
exceeding the observed abundance of GH receptors on arcuate suppresses circulating levels of GH in the rat (496), whereas
GHRH (70,82) and somatostatin expressing neurons (68). For germline deletion of Y1R in mice (Y1R−/− mice) recovers
NPY, expression of the GH receptors is restricted to the ArcN fasting GH release as is seen in NPY−/− mice. Moreover,
(84). GH is thought to only act on select hypothalamic somato- GH release in Y1R−/− mice during the ad libitum fed state
statin and GHRH neurons. Initial reports demonstrated that remains unchanged (225). While suggesting that the Y1R acts
GH treatment activated 11% of GHRH neurons and 5% of as an intermediate between NPY and somatostatin expressing
hypothalamic ArcN somatostatin neurons (69). Subsequent neurons, verification of Y1R expression on somatostatin
observations demonstrated that GH administration induced neurons is yet to be confirmed. The Y1R is widely expressed
cFos expression in only 60% of PeVN somatostatin express- within the periventricular and paraventricular nuclei (266),
ing neurons, whereas cFos expression did not overlap with areas characterized by the abundant expression of somato-
distribution of GHRH neurons in the arcuate nucleus (349). statin mRNA (503), and thus this seems likely. The Y1R com-
By comparison, GH treatment induced cFos expression in monly signals via inhibitory G (Gi) protein-signaling path-
65% of NPY expressing neurons within the ArcN (349), sug- ways, and therefore is anticipated to inactivate somatostatin-
gesting that the NPY system may indeed be a critical inter- expressing neurons. This contradicts the perceived actions
mediate in the regulation of GHRH activity in response to of the Y1R on somatostatin mediated GH output in Y1R–/–
prevailing changes in circulating levels of GH, and thus GH mice. Therefore, should the Y1R promote somatostatin
feedback. While the expression of cFos illustrates activation release, interactions via the Y1R may occur indirectly through
of NPY under these conditions, it should be noted that cFos the inactivation of other neuronal intermediates, or through
expression does not accurately reflect direct interaction with interactions that counter the traditional role of the Y1R. For
NPY neurons, and thus current observations cannot exclude the latter, Y1R mediated activation of neuronal function can
the role of possible intermediates that couple somatostatin occur via non-Gi mediated processed (99).
or GHRH neurons with NPY neurons. Regardless, available As with the Y1R, selective antagonists for Y2R (porcine
data suggest that this may represent a fundamental mechanism NPY 13-36 and Porcine NPY 3-36) inhibit GH secretion
whereby the output of GH to meet physiological demand is (496). Surprisingly, unlike the Y1R, germline loss of the Y2R
coupled to changes in food intake and nutrient availability does not recover GH release in fasting mice, whereas fast-
(485). ing reduces pituitary GH immunoreactivity in wild-type but
In the rat, NPY fibers originating within the ArcN not Y2R−/− mice (301). This suggests that the Y2R partic-
project through the PevN where they surround somatostatin ipates in GHRH mediated GH release, or GHRH mediated
expressing neurons (204). This is corroborated in the sheep GH production. Indeed, assessment of pulsatile GH release
(528) and in the mouse (225), further validating direct in ad libitum fed Y2R−/− mice revealed a significant decline
interactions between NPY and somatostatin neurons through in peak GH release, a derangement in pulse frequency and
synaptic structures. In the mouse, overnight fasting results irregularity, and a reduction in body length (225). Moreover,
in the suppression of pulsatile GH release, occurring almost GHRH neurons express the Y2R (301), whereas antagonism
immediately following the withdrawal of food. This occurs of the Y2R increases electrical activity of GHRH neurons in
alongside an initial elevation in somatostatin and NPY mice (393). It remains unclear whether Y2R-mediated actions
mRNA expression within the ArcN/PevN complex (487), on GH release occur independent of somatostatin expressing
suggesting a potential functional role of NPY in mediating the neurons. NPY neurons do not participate in the regulation of
suppression of GH release following food withdrawal (486). GH pulsatility under fed conditions (225), and thus the Y2R
Indeed, germline deletion of NPY in mice does not impact may contribute to GH release independent of NPY, acting
pulsatile GH release under ad libitum fed conditions, whereas via other factors including PYY. Alternatively, as the Y2R
loss of NPY signaling recovers the suppression of pulsatile inhibits NPY release (62, 87), and deletion of Y2R increases
GH release normally observed in fasting (225). Considering hypothalamic NPY mRNA expression (448), loss of Y2R sig-
well-established species differences wherein food restriction naling in mice may have simply altered the role of NPY in

696 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

mediating GH feedback, thereby impairing GH release. As The action of galanin on GH secretion is thought to occur
with the Y1R, this system remains unexplored, and substan- predominantly within in the central nervous system. Indeed,
tive evidence is needed before a complete understanding of in the rat, galanin stimulates GH secretion after iv or icv
the role of NPY, Y1R, and Y2R in the production and release administration but the effect is stronger after central injec-
of GH can be substantiated. tions (367,394). In addition, immunoneutralization of galanin
Compared to Y1R and Y2R, the role of Y4, Y5, and Y6 in the central nervous system or administration of a GAL-R1
receptors in regulating GH release is almost completely unex- antagonist decreases both GH peak amplitude and frequency
plored. The Y4R subtype (also known as PPYR1) has a greater (161, 319), suggesting that the endogenous peptide partici-
binding affinity for pancreatic polypeptide (199), and thus pates in the regulation of GH rhythmic secretion via activation
may interact with the GH axis independent of NPY. Whilst of GAL-R1.
Y4R deficient mice (Y4R−/− mice) have a lean phenotype, The effect of galanin may be partly due to an increase
recent observations suggest that these receptors likely con- in GHRH release. Indeed, in vitro, galanin is able to induce
trol gonadotropin releasing hormone (GnRH) activity (448), GHRH release from hypothalamic explants or slices (4, 267).
establishing a premise for the regulation of other hypotha- In vivo, passive immunoneutralization with an anti-GHRH
lamic neuronal populations. Loss of Y4R signaling in mice antibody abolishes galanin-induced GH release (366, 367).
contributes to suppressed pituitary GH content in fasting, sug- It remains to be determined whether this effect is direct or
gesting that the Y4R may to some extent directly control GH indirect as GAL-R1 is modestly expressed on arcuate GHRH
release (301). By comparison, Y1 and Y5 receptor subtypes neurons compared to its expression in PeN somatostatin neu-
may act in concert to regulate food intake (129), and thus the rons; 35% of somatostatin neurons in the PeN contain galanin
actions of the Y5R in mediating GH release may be synergis- receptors mRNA and 5% of GHRH neurons in the Arc contain
tic to that of the Y1R. Vasoactive intestinal peptide express- galanin receptors mRNA using double-label in situ hybridiza-
ing neurons in the suprachiasmatic nucleus may participate tion (83). Galanin-induced GH secretion seems to also involve
in the regulation of GHRH activity, an effect that may be somatostatin: galanin-induced GH secretion is blocked by
through Y6R (594). Germline deletion of Y6R in mice results administration of an antisomatostatin antibody (518). A direct
in low levels of hypothalamic GHRH mRNA expression and effect of the peptide on somatostatin neurons is possible since
reduced circulating levels of IGF-1 (594). This suggests that galanin immunoreactive axonal fibers, arising from the ArcN
loss of Y6R expression impairs GH output, and provide com- (146) are closely apposed to somatostatin cell body’s in the
pelling evidence to suggest that Y6R in mice may predict PevN (305) that express GAL-R1 (83).
GH release relative to physiological demand. While we are Neurotransmitters could also be involved in mediating
yet to establish the functional roles of NPY and Y-receptors galanin effects on GH secretion. Anatomical interactions exist
in all mammalian species, it is entirely likely that the NPY between galanin and several hypothalamic neurotransmitters,
systems acts as a critical intermediate between GH output and such as catecholamine or γ-Aminobutyric acid (GABA), and
physiological demand. catecholaminergic and GABA antagonists suppress galanin-
induced GH release (338).
Galanin
Proopiomelanocortin-derived peptides
Galanin is a 29 amino-acid peptide purified in 1983 from pig
intestine (208,521). At the level of the central nervous system, Proopiomelanocortin is a prohormone cleaved into sev-
galanin is present in majority at the level of the hypothalamus, eral peptides that are biologically active: melanotropins
in the ventrolateral portion of the median preoptic area as well (α-, β-, and γ-MSH), endorphins (α-, β-, and γ-endorphins),
as in the paraventricular nucleus, supraoptic, dorsomedial and adrenocorticotropic hormone (ACTH) and corticotrophin-
arcuate nuclei (305, 336, 340, 458). At the level of the ArcN, like-intermediate lobe peptide (CLIP). The last two are mainly
galanin is expressed in a subpopulation of GHRH neurons synthesized in corticotroph cells and melanotroph cells of the
(380) (Fig. 5). pituitary gland, as well as in the gastrointestinal tract. In the
Three different receptor subtypes for galanin have been central nervous system, proopiomelanocortin (POMC) neu-
identified: GAL-R1, GAL-R2 and GAL-R3 (67, 276). They rons are localized in the ArcN of the hypothalamus (Fig. 5)
are 7 transmembrane-domain receptors and present homolo- and in the nucleus of the solitary tract (NTS) (532). The dif-
gies with GPR54 (295). Binding sites are concentrated in the ferent endorphin peptides bind four different opiate receptors
median eminence and hypothalamus (57, 333, 338). Galanin (μ, κ, δ, and γ), closely related to somatostatin receptors.
immunoreactive fibers are detected in the median eminence Opiate receptors mRNA (by in situ hybridization) or binding
(14, 397) suggesting that galanin may exert a direct effect on sites (by radioautography) and receptor proteins (by immuno-
somatotrophs. A stimulatory effect has been reported in vitro histochemistry) distributions vary depending on the receptor
(302,455) but not in all studies (226,394). This effect could be subtype. Whereas μ and k receptors are largely distributed, the
relayed through GAL-R2, which is expressed in the pituitary ∂ subtype is limited to a few brain areas (including the ven-
gland (583). tromedial hypothalamus). In most hypothalamic nuclei and

Volume 6, April 2016 697


Growth Hormone Axis Comprehensive Physiology

median eminence, a high density of κ receptors is observed, with an antisomatostatin antibody (252, 436). Furthermore,
μ and k receptors being even more expressed in the medial icv administration of CRH increases somatostatin concentra-
preoptic area (321, 322, 358). tions in pituitary portal blood (351). CRH could also modulate
In the rat, iv administration of POMC-derived peptides GH secretion by inhibiting GHRH neurons: continuous treat-
like α-MSH (489), β-endorphin (437), and other opiate pep- ment with a CRH antagonist increases the amplitude of GH
tide such as met-enkephalin (66) induces an increase in pulses as well as GHRH expression (360).
plasma GH levels. Accordingly, female melanocortin-3 recep- CRH neurons in the PV nucleus are also direct targets
tor (MC3R) knockout (MC3R−/−) and melanocortin-4 recep- of ArcN NPY neurons as evidenced by anterograde tracing
tor (MC4R) knockout (MC4R−/−) mice exhibit a dimin- (297). To this extent, CRH neurons may serve as a critical
ished responsiveness to the GH-releasing effects of ghrelin intermediates in the regulation of GH output following
(471). However, the role of the melanocortins in the con- changes in food intake, and therefore alterations in NPY
trol of ultradian GH secretion remains controversial. Block- activity.
ade of melanocortin receptors by the nonselective antago- Numerous anatomical data demonstrate a complex inter-
nist, SHU9119, does not modify the activity of the soma- action between several neuropeptides and opioid peptides.
totropic axis (424). Moreover, in Man, obesity due to MC4R For example, galanin and POMC ArcN neurons are inter-
deficiency is associated with increased linear growth and connected (220) and NPY-containing fibers project to β-
final height, fasting hyperinsulinemia, and incompletely sup- endorphin positive neurons that send synapses onto dopamin-
pressed growth hormone secretion (328). Concerning opioid- ergic neurons (221, 222). An additional level of complexity
POMC-derived peptides, Naloxone, a μ receptor antagonist, is represented by the numerous colocalizations that occur in
decreases circulating GH (66) or it does not alter parame- ArcN (207). Moreover, the degree of colocalization of neu-
ters of GH secretion (515, 559). Despite high concentrations ropeptides in GHRH or somatostatin neurons varies accord-
of ß-endorphin in the portal blood (306), opioid peptides do ing to adiposity (232), stressing the relationship between
not act directly on the somatotroph cell (437) but may rather metabolism and GH secretion. This is reviewed in the fol-
act through modulation of GHRH and somatostatin neurons. lowing section.
GH response to an administration of opiates is consider-
ably reduced after selective destruction of GHRH neurons
in the ArcN by monosodium glutamate (13) or immunoneu- Integration between Regulators of Food
tralization of endogenous GHRH (344, 563). Alternatively, Intake/Metabolism and GH Release
the increase in GH secretion after administration of μ opioid
receptors agonists in the PevN (578) is in favor of an inhibitory The production and release of GH changes considerably rela-
effect of opiate peptides on somatostatin release. Opiate pep- tive to food intake and body composition. Therefore, it is not
tides are indeed able to inhibit somatostatin release induced by surprising that a number of intermediates that are traditionally
potassium depolarization from hypothalamic explants (128). seen as regulators of food intake and metabolic balance con-
The existence of anatomical connections between POMC tribute to the short- and long-term regulation of GH release.
and GHRH neurons on one side, POMC and somatostatin Of these, discovery and the identification of the actions of
neurons on the other side, suggests that POMC neurons could ghrelin (a potent orexigenic factor) highlighted an additional
regulate GH secretion by connecting ArcN GHRH neurons level of control, whereby traditional regulators of GH pulsatil-
and PeVN somatostatin neurons. Indeed, in the ArcN, β- ity (somatostatin and GHRH) are no longer seen as the only
endorphin immunoreactive fibers were shown in close appo- modulators of GH pulsatility. Rather, it is now accepted that
sition to GHRH cell bodies (112) and to somatostatin cell GH output is largely modified by metabolic needs, and thus is
bodies in the PevN (146). Reciprocally, binding sites that are under feedback control of numerous intermediates that signal
sensitive to octreotide, a somatostatin-receptor 2 preferring hunger, satiety, or excess or under nutrition. Indeed, many of
agonist, are present on POMC positive neurons (145, 147), the mechanisms that modify GH output relative to nutrient
suggesting that somatostatin neurons also modulate the activ- supply are yet to be defined. To this extent, it is not yet possi-
ity of POMC neurons. ble to provide an exhaustive list of all factors that modify GH
output relative to nutritional or metabolic demand. Therefore,
this section will predominantly focus on ghrelin and ghrelin
Corticotropin-releasing hormone
derived peptides, and an only a brief summary of other regu-
Corticotropin-releasing hormone is a 41 amino-acid peptide lators (including insulin, free fatty acids and glucose) will be
(477) synthesized in various regions within the central ner- provided.
vous system. CRH is highly expressed in the paraventricular
nucleus (498) (Fig. 5). CRH decreases basal GH secretion in Ghrelin
the rat after icv injection (252, 391, 436) and GHRH-induced
GH secretagogues and ghrelin discovery
GH release in humans (24). This effect seems to be relayed
through an increase in somatostatin release as the inhibitory In their search for compounds able to mimic or amplify
effect of CRH on GH release is abolished by a treatment the endogenous secretion of GH, Bowers and collaborators

698 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

directed their attention to opiates known to stimulate GH


and PRL secretion. In doing so, they synthesized a derivative
from Met-enkephalin, GHRP-6 (His-D-Trp-Ala-Trp-D-Phe-
Lys-NH2), devoid of opiate activity. GHRP-6 was the first
synthetic GH secretagogue (GHS) described to have activ-
ity in vivo without binding any receptor known to stimulate
GH secretion. During the subsequent 20 years, compounds
that are smaller, more stable, and soluble have been devel-
oped and are either peptides (54, 123) or non-peptides such
as L692-429, L-692-585, or MK-0677 (88, 89, 404). A spe-
cific receptor for these synthetic compounds, the GH secreta-
gogues receptor (GHS-R), was cloned from porcine pituitary
in 1996 (223). The effects of GHS on GH release have been
described in humans as well as in different animal species
such as the sheep (183), the guinea pig (138, 139) and the rat
(93, 100, 586). The effects of GHS are not completely spe-
cific of GH secretion, as some have reported an activation of
the hypothalamic-pituitary-adrenal (HPA) axis by GHRP-6
for example (526). In humans, GHS stimulate GH, prolactin,
ACTH and cortisol (157). Discovery and characterization of
GHS and the GHS-R lead to the eventual identification of the
potent endogenous GH agonist, ghrelin.

Figure 6 Ghrelin acts within the hypothalamus and the anterior pitu-
itary gland indirectly (through inhibiting somatostatin- and stimulat-
Ghrelin: structure and function ing GHRH-neuronal activity), and directly by stimulating somatotrophs,
respectively. The actions of ghrelin are mediated through select acti-
In 1999, ghrelin was isolated as the endogenous ligand for vation of the GHS-R. Actions of ghrelin on GH release may involve
the GHS-R1a (273, 531) and for its ability to stimulate GH interactions with feeding circuits, specifically through neuropeptide-Y
secretion (273,274). A cDNA, identical to preproghrelin, was (NPY)-expressing neurons located within the arcuate nucleus (ArcN).
Stimulatory interactions are highlighted in green arrows. Inhibitory
cloned independently and named prepro-Motilin Related Pep- interactions are highlighted in orange arrows. Additional abbrevia-
tide (preproMTLRP) (531). Ghrelin is a 28 AA peptide that tions: ME, median eminence; PevN, periventricular nucleus; PvN, par-
is posttranslationally modified with an eight-carbon fatty acid aventricular nucleus; POMC, pro-opiomelanocortin; GAL, galanin.
on a serine in position 3 by the enzyme Ghrelin-O-Acyl-
Transferase (GOAT) (185, 592). The acylation of ghrelin is
required to activate the GHS-R and to mediate its effects
Ghrelin-induced GH release from somatotrophs is
on GH secretion and food intake. Indeed, acylated ghrelin
mediated through increased intracellular Ca2+ levels via IP3
potently stimulates GH secretion both in vitro and in vivo in
signaling pathway [reviewed in (262)]. GH response to GH
several animal species, including rodents (530), non-human
secretagogues is only partially dependent on the GHRH sig-
primates (263) and humans (63, 502, 549). In humans, con-
naling, as the response to acutely injected GHRP-2 is severely
tinuous infusion of ghrelin during a 24-h period stimulates
reduced but not completely blunted in lit/lit mice bearing a
pulsatile (burst mass as well as frequency) and entropic (the
mutation in the GHRH-R (409). In addition to a direct stimu-
GH secretory pattern becomes more irregular) modes of GH
latory effect on the pituitary, ghrelin amplifies GH secretory
secretion (549).
pattern by indirect hypothalamic actions. In rat hypothalamic
slices in vitro, 85% of ArcN neurons projecting to the median
eminence are excited by ghrelin and inhibited by somatostatin
Ghrelin: mechanism of action
(356). The expression of the GHS-R1a in 25% of ArcN
The GHS-R is highly expressed in the pituitary gland as GHRH neurons suggests that the effect of ghrelin is direct
well as in the hypothalamus (178, 181, 361, 593), suggest- on GHRH neurons (512, 575). Consistent with these neu-
ing both direct and indirect mechanism of action (Fig. 6). roanatomical observations, ghrelin stimulatory action on GH
Using a GHSR-eGFP reporter mouse line, it was found that is partly mediated via a modulation of the activity of GHRH
77% of somatotrophs express GHS-R-eGFP in both males neurons, as it disappears after passive immunization with
and females (428). In the same mouse line, eGFP expression an anti-GHRH antiserum (508), and ghrelin can stimulate
is low in several midbrain regions and in several hypothala- GHRH release from hypothalamic explants (582). In addi-
mic nuclei, particularly the ArcN, where robust GHSR mRNA tion, in GHRH-eGFP mice, ghrelin increases GHRH neurons
expression is well characterized (512,575); thereby question- excitability by increasing their firing rate (393). Although
ing the model (320). initially reported as purely postsynaptic, others demonstrated

Volume 6, April 2016 699


Growth Hormone Axis Comprehensive Physiology

that ghrelin decreases GABA-ergic synaptic transmission food intake (600). However, when administered at high doses,
in 44% of recorded GHRH neurons, an effect blocked by BIM-28163 displayed an agonist activity in addition to its
a selective GHS-R antagonist, while it did not involve glu- antagonist properties as it increased food intake and weight
tamatergic neurotransmission (141). Unlike GHRH, ghrelin gain as well as cFos activity in the dorsomedial nucleus of the
seems refractory to the inhibitory action of somatostatin. hypothalamus (189, 194). These effects could be mediated
Indeed, ghrelin stimulates GH secretion when administered through actions on a receptor distinct from the GHS-R1a.
intravenously during either low (peak GH) or high (trough Taken together, these data suggest that endogenous ghrelin,
GH) endogenous somatostatin tone in the rat (530). However, via its action on the GHS-R1a, amplifies the pulsatile pattern
icv injection of ghrelin at trough times does not significantly of GH secretion but the mechanisms by which endogenous
increase plasma GH levels (508); even though ghrelin inhibits ghrelin modifies food intake remain to be defined and may
somatostatin release from perifused rat hypothalamus (530). involve a novel receptor.
In prepubertal sheep, ghrelin infusion for 6 hours during The difficulty to interpret data from pharmacological
3 consecutive days increases accumulation of somatostatin manipulations of the GHS-R1a is further complicated by;
stores in the PevN perikarya and in the terminals of the (i) the fact that this receptor is associated with multiple signal
median eminence, resulting in enhanced GH release (416), transduction pathways (72,212), (ii) the existence of a variety
suggesting that ghrelin impairs somatostatin release in this of ligands or cofactors in addition to ghrelin, like adenosine,
species as well. that can interact with the GHS-R at different binding sites
In addition to its primary effect as a GH secretagogue, (474) or that can behave as positive or negative modulators of
ghrelin exerts a powerful orexigenic effect after acute injec- endogenous ghrelin signaling (406), and (iii) the existence of
tion (533), primarily mediated through NPY/Agouti-related constitutive activity for the GHS-R (212, 213, 215, 216, 400).
protein-expressing neurons in the ArcN [GHS-R is highly
expressed in this population of neurons (373)]. These neu-
Genetics
rons modulate GH secretion through their connections to
somatostatin-expressing neurons in the PevN (225) provid- Transgenic mice in which ghrelin-producing cells develop
ing a potential intermediate site of action in the regulation of into ghrelinoma with elevated plasma ghrelin levels exhibit
ghrelin-mediated GH release. elevated IGF-1 levels and display increased GH response
The vagus nerve is a mediator of ghrelin-induced GH to GHRH, although they have reduced appetite and body
secretion and is necessary for the full GH releasing activity weight, suggesting that chronic elevation of ghrelin activates
of ghrelin. Indeed, animals with a disruption of vagal con- the GH/IGF-1 axis (236). In contrast, adult preproghrelin
nections display a decreased GH response to peripheral and knockout (ghrl−/−) mice have no major weight or growth
central ghrelin injections (7). Interestingly, blockade of the alterations under balanced feeding environment (494). In
peripheral cannabinoid-1 receptor subtype with rimonabant young ghrl−/− mice, the amplitude of GH secretory bursts is
reduces ghrelin-induced GH secretion but does not modify reduced compared to wild-type littermates whereas the secre-
GHRH-induced GH secretion, whereas vagotomy suppresses tory pattern is not significantly altered (195). This suggests
the inhibitory action of rimonabant on GH. This suggests that that endogenous ghrelin acts as an amplifier of GH pulsatility.
the endocannabinoid system relays the effects of ghrelin on In contrast, GHS-R knockout mice (Ghsr−/−) present with
GH secretion by the vagus nerve (6). mildly lower body weights and reduced IGF-1 levels (495),
supporting a role of the GHS-R in growth. Possible compen-
satory mechanisms due to the absence of ghrelin during the
Pharmacology
developmental period to explain the lack of strong phenotype
Several GHS-R antagonists have been developed to decipher in ghrl−/− mice have been ruled out. Indeed, despite induc-
the role of the ghrelin/GHS-R pathway in physiological condi- tion of a postweaning reduction in circulating ghrelin levels
tions. Repeated injections of the antagonist [d-Lys3]-GHRP6, in mice expressing human diphtheria toxin receptor (DTR)
an analog of one of the synthetic GHS (18), does not modify in ghrelin-secreting cells and treated with DTR, no evidence
ultradian GH secretion (388) although it suppresses feeding of growth retardation was observed: body weight, nasoanal
in mice after acute or chronic icv injections (18). Results length and IGF-1 plasma levels were unmodified. Pulsatile
obtained with this compound are difficult to interpret because GH secretion was not measured in these mice and a reduced
it also binds melanocortin receptors (459). Another developed GH response to GHRH was only observed in 5-week-old
molecule, JMV2810, antagonizes the effects of hexarelin (a males but not females (15). To this extent, different genetic
synthetic ghrelin agonist) on food intake but not on GH secre- mouse lines targeting the various forms of ghrelin or ghrelin
tion after acute injection (126) but JMV2810 effects per se receptors have only added to the array of questions regarding
on spontaneous GH secretion or food intake were not inves- the true endogenous role of ghrelin in regulating GH pulsatil-
tigated. Subcutaneous or central infusion of another selective ity, and the actions of GH on body composition and growth.
GHS-R antagonist, BIM-28163, during 48 h-administration Constitutive activity of the ghrelin receptor could pro-
through an osmotic mini-pump in rats decreased the amplitude vide a residual signaling in mice lacking ghrelin (412). Phys-
of GH secretory peaks by 46% without significantly affecting iological significance supporting a link between GHS-R1a

700 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

constitutive activity and the activity of the GH/IGF-1 axis (0.29 ± 0.08) further suggesting a divergent role for ghrelin
is substantiated by human genetic studies. Interestingly, in in the control of GH release relative to nutrient supply. This
humans, rare natural mutations of the GHS-R, transmitted is corroborated in mouse studies, wherein the suppression of
on a dominant or a recessive mode, are associated with short GH release in the mouse during fasting does not coincide with
stature and/or GH deficiency (400,401). Furthermore, the dis- altered acylated ghrelin release (487), whereas the eventual
ease mechanism shared by the very few cases documented so rise in circulating levels of ghrelin during a prolonged fast
far is a complete or a partial loss of constitutive activity of does not correspond to an increase in GH output (486).
the mutant GHSR1a receptors (A204E, F279L, and R237W)
(215, 400, 560).
Other ghrelin-derived peptides
As stated earlier, the activity of ghrelin to stimulate GH
secretion and food intake depends on the presence of an eight- The preproghrelin gene encodes other functional or nonfunc-
carbon fatty acid residue attached to Ser in position 3. Mice tional molecules in addition to acylated ghrelin, including des-
invalidated for the GOAT gene (Goat−/− mice), encoding the acylated ghrelin and obestatin, or other C-terminal proghrelin
enzyme that acylates ghrelin, are currently the most appropri- peptides whose physiological significance still needs to be
ate model to investigate specifically the role of acylated ghre- clarified (466). To this extent, a lack of ghrelin in preproghre-
lin in the regulation of GH secretion and feeding, as circulating lin deficient animals might be counterbalanced by the lack of
acylated ghrelin secretion is totally absent in this model (598). other proghrelin-derived molecules with postulated antago-
In contrast to Ghsr−/− mice, growth retardation or changes nistic actions.
in body composition have not been reported in Goat−/− mice Des-acylated ghrelin is the most abundant form in
(598). A moderate reduction in weight was only observed plasma and accounts for up to 80% to 90% of total ghrelin
when mice were fed an octanoate rich diet (264). As observed (274). Although nonendocrine biological activities have been
in ghrsl−/− or Ghsr−/− mice, food consumption is not altered attributed to the binding of des-acylated ghrelin to an uniden-
in Goat−/− mice. Current observations from Goat−/− mice tified receptor, recent studies suggest that it also modulates the
show derangements in peak GH output, specific to young effects of acylated ghrelin (231). However, it does not bind to
age, and an overall rise in GH pulse frequency and irregular- the GHS-R and there currently is no pharmacological tool to
ity (584). In this context, the acylated form of ghrelin may examine its biological role. At any rate, this ghrelin variant is
contribute to peak GH output, while entraining mechanisms inactive on GH secretion and studies investigating its effects
of patterned GH release. It remains unclear why this derange- on feeding have not been conclusive (17, 230). Interestingly,
ment in GH output does not contribute to clearly discernable transgenic mice overexpressing des-acylated ghrelin had sig-
differences in body length. nificantly reduced body weight, reduced food intake and body
One important function of GH is to regulate glucose fat mass and moderately decreased linear growth compared to
and lipid metabolism as it induces hyperglycemia and elicits nontransgenic mice at 44 weeks of age (17). These data sug-
changes in fat distribution and mobilization. When submit- gest that des-acylated ghrelin modulates, through an unchar-
ted to a 60% caloric restriction, young Goat−/− mice have acterized pathway, growth and metabolism.
reduced GH secretion and are unable to maintain normal blood Obestatin, (contraction of “obese” and “statin” denot-
glucose (598). Thus, acylated ghrelin, although not essential ing suppression) is another bioactive 23 amino-acid pep-
for normal growth, may have an essential function in mice tide derived from the C-terminal portion of the 117 AA
during adulthood by elevating GH levels during severe caloric preproghrelin precursor. It was originally proposed as the
restriction, thereby maintaining fasting blood glucose levels endogenous ligand for GPR-39 (332, 597). Obestatin was
and allowing survival. initially shown to decrease appetite and weight gain thus
opposing ghrelin effects but subsequent studies failed to con-
firm such effects or that obestatin was the cognate ligand
Relationship between ultradian ghrelin and GH for GPR39 (86, 214, 292). Obestatin was also described as a
secretion
functional antagonist of ghrelin actions in rodents (332, 597).
Growth hormone and ghrelin secretory patterns were not Physiological effects of obestatin on GH are not consistent
strictly correlated in two studies measuring total or acy- from one study to another. Whereas some studies do not
lated ghrelin circulating levels in freely behaving rats sampled observe any effect of obestatin per se after acute injections
every 20 min (529, 601). Accordingly, GH secretory pattern in rats (601), other studies report that obestatin decreases
(peak frequency or number) is not modified after treatment GH secretion in vivo and plasma IGF-1 levels after chronic
with a GHS-R1a antagonist (600) or in ghrl−/− mice (195). treatment in mice and non-human primates (312). In rodents,
In contrast, in humans, using a higher sampling frequency obestatin antagonizes GH secretion and food intake induced
and very selective sandwich enzyme-linked immunosorbent by ghrelin only when ghrelin and obestatin are administered
assays (377), a clear relationship between acyl ghrelin and in equimolar quantities (196, 312, 381, 601). A direct action
GH secretion was observed (mean of the individual peak cor- of obestatin on somatotrophs has not been clearly validated.
relations 0.70 ± 0.04). This was only seen in subjects given In pituitary cell cultures from non-human primates (baboon)
standardized meals and correlation dropped in fasted subjects and mice, obestatin inhibits GH expression and release after

Volume 6, April 2016 701


Growth Hormone Axis Comprehensive Physiology

24 h of incubation and inhibits ghrelin-induced GH secretion, discovery of ghrelin (273), and the identification of neuronal
suggesting a pituitary site of action. This is associated with mechanisms that couple GH output with food intake.
a reduction in the expression of pituitary GHRH-R, SST1, Chronic actions of GH counter insulin function, resulting
and SST2 (312). However, earlier studies in rat perifused in peripheral insulin insensitivity, and insulin hypersecretion
pituitaries did not detect a direct action at the pituitary level [reviewed in (555)]. The well-defined anti-insulin actions of
(597, 601). It is possible that ghrelin and obestatin interact GH may underscore the progressive decline in GH release
pharmacologically at the hypothalamic level to modulate food resulting in GH deficiency that is commonly seen in obe-
intake and GH secretion. Obestatin treatment causes a reduc- sity (375, 426, 548). Indeed, the progressive decline in GH
tion in hypothalamic GHRH expression (312). In addition, secretion with weight gain correlates with a rise in cir-
obestatin antagonizes ghrelin-induced cFos activation in the culating levels of insulin in both humans (550) and mice
arcuate nucleus and antagonizes ghrelin-induced inhibition of (488). This suggests that GH release is tightly controlled by
GABA synaptic transmission onto GHRH neurons (141,195). prolonged changes in energy intake, and potentially insulin
Concerning the importance of preproghrelin derived pep- release. Indices of this inverse relationship exist across many
tides in terms of phylogeny, it should be noted that genes metabolic states. In humans, fasting (227), malnutrition result-
encoding ghrelin and GOAT are missing in the platypus ing in anorexia (345), and insulin-dependent type-1 diabetes
genome, whilst, as reported in other species, the GHS-R is mellitus (10, 19, 191, 197) are associated with decreased lev-
present and expressed in brain, pancreas, kidney, intestine, els of insulin and a rise in GH release, whereas hyperphagia
heart, and stomach (198). (106,107) and obesity (375,426,548) are associated with ele-
vations in circulating measures of insulin and a fall in GH
release. Given this well-established relationship, it comes as
Insulin a surprise that few observations exist to establish a functional
In both humans and animals, acute and chronic GH treatment relationship whereby insulin may define GH release.
differentially impacts carbohydrate and lipid metabolism. Anecdotal evidence from humans suggests a potential
Acute effects of GH are thought to be insulin-like, direct link between hypoinsulinemia and GH excess. Inten-
resulting in a decrease in blood glucose, the stimulation of sive insulin treatment in type-1-diabetic patients reverses GH
muscle glucose uptake, and an increase in adipose-specific hypersecretion (19), whereas calorie restriction to reverse
glucose transport and lipogenesis (114, 354). Provocation hyperinsulinemia (259) results in the recovery of GH secre-
of GH release in humans through the administration of tion, presumably in response to a reduction in insulin levels
insulin (insulin-induced hypoglycemia) is often used to assess (119). Evidence that define the relationship between GH and
GH-axis function (153). The insulin-mediated GH response insulin secretion almost always document opposing changes
observed in humans most likely occurs in response to the acute in GH insulin release, whereas attempts to disrupt this balance
fall in glucose levels (445), and thus not through direct insulin to define the physiological context of inverse GH/insulin bal-
action. This glucose-dependence is further demonstrated by ance are scarce. Overeating in humans suppresses GH release
the inhibition of GH and GHRH-induced GH release follow- (106, 107). This occurs before observable changes in weight
ing an acute rise in blood glucose levels (commonly seen dur- gain, and alongside an elevation of 24-h insulin secretion. GH
ing oral or intravenous glucose loading) (115, 166, 470). Crit- treatment at this time (to reverse the suppression of endoge-
ically, plasma GH levels in other species following insulin- nous levels of GH) results in a further increase in mean daily
induced hypoglycemia does not reflect increased GH release, circulating levels of insulin, fasting insulin, and the insulin
with measures from pigs, dogs, and cats showing no or a response to a meal (106). In this context, the physiological
modest change in GH output in response to insulin induced suppression of GH during sustained periods of calorie excess
hypoglycemia (275,315,534). Insulin-induced hypoglycemia would facilitate insulin action (106). This is in line with the
in sheep promotes the release of somatostatin into portal established role of GH as a regulator of insulin sensitivity.
circulation, culminating in the eventual suppression of GH Importantly, these measures also highlight the existence of a
release (159). Within the rats and rabbits, GH release is sup- potential feedback-mechanism whereby insulin may mediate
pressed (331, 501). Since the mean period of the GH rhythm GH release.
was not significantly altered by hyperglycemia—although the Insulin inhibits GH synthesis from isolated pituitary cells
amplitude of the pulses of half of the hyperglycemic rats was by inhibiting GH gene expression (588, 589). Insulin acts
markedly depressed—it was initially concluded that insulin selectively on insulin receptors within the pituitary gland
does not significantly affect the endogenous ultradian rhythm (314) to suppress GH release from isolated somatotrophs
of GH secretion in the rat (514). Similar reports exist in (314,334). These effects persist regardless of the development
Man, with data showing no direct correlation in immediate of systemic insulin resistance (314), suggesting that soma-
peak GH release relative to circulating glucose or insulin totrophs remain insulin responsive in obesity. Thus, insulin
levels (71,420). Moreover, early reports failed to consistently may modulate GH release relative to weight gain, and promote
demonstrate altered postprandial GH output specific to a meal the sustained suppression of GH release in obesity, regardless
(8,22), suggesting that GH release may not be coupled to food of systemic insulin resistance. Targeted in vivo deletion of
intake. This premise was significantly revised following the insulin receptors from somatotrophs results in an increase

702 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

in pituitary GH content and release (163). Based on selec- release that is seen following acute treatment with acipimox
tive inactivation of the Insr and IgfIr genes in mouse soma- may also occur as a consequence of altered insulin feedback.
totrophs, it can be concluded that the suppressive actions of Acipimox reduces circulating levels of insulin in obese sub-
insulin on GH release occur independent of IGF-1 (163). It jects (12, 418), and thus the reduction of GH secretion as
is thus likely that insulin levels predict GH release indepen- observed in obesity may simply reflect prevailing levels of
dent of IGF-1, and that insulin may provide critical feedback insulin.
to alter GH release relative to long-term metabolic require- In humans, the augmented secretion of GH during fasting
ments. It should be noted that loss of somatotroph-specific is thought to stimulate the release of FFA (193, 205). Indeed,
insulin receptor expression does not completely reverse the reversal of the fasting induced rise in GH release in humans
suppression of GH release seen during dietary induced weight impairs whole body lipolysis, as measured through a partial
gain and obesity (163), and therefore insulin may not solely reduction in glycerol Ra (the rate of appearance of glycerol, an
predict GH release in weight gain and obesity. Alternatively, index of lipolysis) following antagonism of GH output during
the actions of insulin on GH release may not be restricted to a 24 and 48 hour fast (449). Of interest, these observations
the anterior pituitary gland. Insulin receptors are expressed further demonstrate that, while contributing to the rise in FFA
throughout the hypothalamus, including the ArcN and PevN release, factors other than GH predominantly promote FFA
(148, 201, 249, 572); however, a comprehensive assessment release in fasted humans. Interestingly, in fasted mice GH does
of the role of insulin in regulating hypothalamic control of not mediate the rise in FFA release. Rather, a rapid elevation
GH output is needed. Thus, while insulin may modulate GH in FFA release following food withdrawal occurs alongside
release acting through the hypothalamus, this premise remains the suppression of GH secretion. In addition, the elevation
completely unexplored. As insulin does not predict ultra- in circulating levels of FFAs in response to food deprivation
dian patterns of GH release (discussed earlier), the actions persists in GH receptor knockout mice (487). This suggests
of insulin within the hypothalamus to mediate GH output that, unlike in humans, GH does not contribute to the early
may be limited to the infradian regulation of peak GH output. rise of FFA in mice in response to fasting. Species difference
should therefore be considered in future studies regarding the
relationship between GH and FFA during fasting conditions.
Free fatty acids Although there are still many unanswered questions, it is
An important metabolic effect of GH is to drive the release of generally believed that circulating FFA levels are negatively
free fatty acid (FFA) from adipose tissues (193,205), whereas correlated with GH levels in all species, mainly through mod-
FFAs in turn may exert feedback on GH release (the physi- ification of pituitary response to hypothalamic GHRH and/or
ological context of this remains to be confirmed). Studies in somatostatin signals.
humans and animals using a lipid infusion approach revealed
a direct inhibitory effect of FFA on GH secretion, presumably
through the direct suppression of GH release from soma- Leptin
totrophs and an indirect inhibition of somatotrophs at the Leptin is a hormone secreted from adipocytes in proportion
hypothalamic level (9, 61, 74). Interestingly, in obesity, an to fat mass (154, 316). By inhibiting appetite and promoting
increase in circulating levels of FFA coincides with a decrease energy utilization, leptin plays an important role in main-
in GH secretion, and the pharmacological suppression of FFA taining energy homeostasis (243). The effect of leptin on
by the antilipolytic drug acipimox is associated with a recov- regulating GH secretion is well established; however, some
ery of GH secretion in obese subjects (12,105,374,418). Thus, debate persists regarding potential inhibitory and stimulatory
it was proposed that impairment of GH secretion observed in actions.
obesity may occur as a consequence of an elevation in circu- In vitro studies support a direct stimulatory role of leptin
lating levels of FFA (171). However, a recent meta-analysis on GH release using pig pituitary slices alone or coincubated
has demonstrated relatively stable circulating levels of FFA with the stalk median eminence (2, 450). However, a direct
regardless of the severity of obesity (250), and thus it remains inhibitory effect of leptin on ovine pituitary somatotrophs is
unknown whether obesity-associated alterations in circulat- also well documented (439, 440), with a decrease in GHRH-
ing levels of FFAs contribute to altered GH output. In agree- stimulated GH secretion from leptin-treated primary cultured
ment with this, a reduction of GH secretion following dietary- cells. Such inhibitory effect may depend on the nutritional
induced body weight gain does occur in mice without an eleva- status of the animals (265). In vivo, central infusion of leptin
tion of FFA concentrations (224). Therefore, the impairment promotes GH secretion by acting concurrently at GHRH and
of GH secretion in the early stage of obesity may not occur in somatostatin neurons in rodents (96, 510, 561), and the fast-
response to an elevation of FFA, at least directly. Rather, given ing induced fall in GH secretion in rats is fully prevented by
that elevated levels of FFA are associated with insulin resis- central leptin administration (288). In addition, somatotroph
tance and the hypersecretion of insulin (242), decreased GH specific deletion of leptin receptors results in a reduction in
secretion in obesity may occur as a consequence of altered GH immunopositive cell numbers and an overall reduction in
insulin feedback, rather than in response to an elevation in GH secretion (90). Thus, leptin appears to maintain GH secre-
circulating levels of FFAs. To this extent, the recovery of GH tion in a normal range while mediating GH output relative to

Volume 6, April 2016 703


Growth Hormone Axis Comprehensive Physiology

nutrient supply. The mechanisms of action through which animals. Species difference exists clearly between laboratory
leptin modulates GH release in the rodent remain unknown, animals (mainly rodents) and humans. In this section, we will
and may require interactions with neuronal intermediates. For briefly discuss existing data on rodents and humans, focus-
example, leptin modifies NPY-expressing neuronal activity ing on GH levels under hyper- or hypoglycemic conditions.
[specific to food-intake (479)], whereas NPY-neurons are crit- Attempts for in vivo assessment of GH output relative to cir-
ically involved in the regulation of GH release. In this context, culating levels of glucose are compromised by accompanying
leptin infusion in the rodent may prevent the rise in NPY activ- changes in circulating levels in insulin. As previously dis-
ity that is normally associated with fasting, thereby preventing cussed, insulin is a key regulator of GH release, and thus
NPY-mediated suppression of GH release. However, the bio- must be considered alongside attempts to define the actions
logical significance of this interaction (considering species of glucose on GH release.
differences) remains completely unexplored in humans.
Leptin may act as an adipostat, signaling nutrient status Hypoglycemia
to the hypothalamus, thereby mediating long-term changes
The regulation of GH secretion during hypoglycemia may
in hypothalamic control of hormone output. During periods
occur at the level of the pituitary gland, and/or hypothalamus
of excess, such as obesity, an increase in circulating levels
through interactions with GHRH and somatostatin neurons.
of leptin is observed alongside a decrease in GH secretion
Since brain tissues use almost exclusively glucose to provide
(352, 488). This can be attributed to an impairment of lep-
energy (395), determination and regulation of glucose in cir-
tin signaling and/or the development of leptin resistance due
culation becomes essential in keeping normal brain function.
to severe obesity (369, 599). Conversely, administration of
Moreover, glucose-sensing neurons contribute to the appro-
leptin in leptin deficient ob/ob mice restores impaired GH
priate physiological responses to maintain peripheral glucose
secretion (313). However, in other obesity models, prevailing
levels within a normal range. To this extent, GHRH neurons
elevated endogenous leptin levels makes it irrational to add
may contribute to peripheral glucose homeostasis. Through
exogenous leptin. In humans, congenital absence of leptin or
over expression of glucokinase (a critical enzyme that facil-
mutations of the leptin receptor result in a reduction in GH
itates phosphorylation of glucose to glucose-6-phosphate)
secretion (95) and a blunted GH response to GH stimuli (396).
in EGFP-tagged ribosomal protein construct mice, it was
In this context, it is more than likely that other inhibitory fac-
demonstrated that hypoglycemia activates GHRH neurons
tors (including insulin) may override the stimulatory effect
(478). It is important to note, however, that hypoglycemia
of leptin and ultimately contribute to the suppression in GH
in mice causes a reduction in GH release (486,487), a process
secretion in obesity.
thought to be mediated by selective activation of NPY neurons
Fasting results in a decline in leptin levels out of pro-
(225) Therefore, the role of direct glucose sensing in GHRH
portion to the reduction of body fat mass (51, 568). Inter-
neurons, in combination with the effect of endogenous sys-
estingly, administration of recombinant methanoyl human
tems that may modulate GH release specific to hypoglycemia
leptin (r-metHuLeptin) to normal weight subjects following
are yet to be defined. Herein, neuronal integration with other
72 h of fasting (to reverse the fasting associated hypolepti-
systems, including NPY expressing neurons, may override
naemia) (80) or to hypothalamic amenorrhea women (with
GHRH responses. See above (page 9-10) for a more com-
chronic energy deficit and relative leptin deficiency) does
prehensive overview of the role of NPY in mediating GH
not alter GH secretion (81). Thus, unlike rodents whereby
output.
administration of leptin restores suppressed GH secretion
following food deprivation (288), leptin deficiency may not
Hyperglycemia
serve a direct inhibitory signal on GH secretion during long-
term fasting in humans. It is worth mentioning that fasting- In diabetic rat models, either type I or type II, depressed GH
induced immediate change in GH secretion is contradictory release and blunted GH response to GHRH were reported
between rodents (decrease) and humans (increase), therefore, (48, 239, 378). Hyperglycemia inhibits GH secretion through
species difference in the role of leptin in mediating GH secre- both direct and indirect mechanisms, by a direct action
tion should not be ignored. Nevertheless, it should be noted on pituitary somatotrophs to blunt GH release in response
that r-metHuLeptin replacement does increase the starvation- to GHRH stimulation (389), or by hyperglycemia-induced
induced decrease of total IGF-I (a major by-product regulated hyperinsulinemia and hypersomatostatinemia (405). Hyper-
by GH) in humans (81). Thus, the role of leptin in regulating glycemia causes elevated plasma levels of somatostatin-like
IGF-1 independent of GH requires further investigation. immunoreactivity (405) and administration of somatostatin
antiserum to streptozotocin-induced diabetic rats resulted in
an increase in GH plasma level (506). Such results also
Glucose indicate that pituitary somatotroph responses to somato-
Generally, elevated glucose levels in plasma (hyperglycemia) statin may also be involved. In isolated pituitary cells from
occurs alongside a reduction in basal and pulsatile GH secre- streptozotocin-induced diabetic rats, GHRH induced GH
tion, whereas reduced glucose levels (hypoglycemia) occurs secretion is significantly reduced (389). The GH content of
alongside an increase in GH release in humans, and in large pituitaries and GHRH content of hypothalamus from diabetic

704 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

rats are also significantly decreased compared to control rats will be discussed when references to GH in Man are not avail-
(389). Growth hormone releasing hormone receptor levels able, or when mechanistic insights were obtained from other
in pituitary glands have been evaluated in streptozotocin- species. In this regard it is important to note that while the
induced diabetic rats and the increase of the 2.5-kb GHRH-R pulsatile appearance of GH in circulation is conserved across
mRNA transcript coincided with a decrease of hypothalamic all mammals characterized to date, the amount of GH release,
GHRH mRNA levels (33). Overall, the depressed GH secre- and patterns of GH output across species differ considerably.
tion induced by hyperglycemia may result from a combina- This may be due to differential sampling practices (i.e., dif-
tion of decreased GH content in the pituitary gland, increased fering sample intervals, assay sensitivity, and statistical anal-
release of somatostatin in hypothalamus. However, given the ysis) or may reflect underlying biological properties of GH
anticipated actions of insulin, elevated insulin secretion within release that are specific to each species. Indeed, differences in
these models may account for reduced pituitary-sensitivity to GH output between small and large mammals are thought to
GHRH stimulation, and diminished GHRH synthesis. occur as a consequence of differing metabolic needs. We will
In human diabetes, GH secretion shows different charac- not provide a comprehensive overview of GH output across
teristics according to different stages and types of diabetes. species, as this does not directly contribute to the general
In type I diabetes without significant obesity, pulsatile GH aims of this article. A summary of GH output in Man and in
secretion is increased (19, 197). This occurs alongside an different mammalian species is provided in Tables 1 and 2,
increase in somatotroph sensitivity to GHRH (414). This dif- respectively. For this, observations were limited to publica-
fers from type I diabetic rat models, showing a remarkable tions wherein measures of pulse irregularity [i.e., approximate
species difference. The increase in sensitivity of somatotrophs entropy (ApEn)] were provided.
to GHRH might be caused by a decrease in somatostatin out-
put from hypothalamus to pituitary gland (234). In type II
diabetes with obesity, GH levels are decreased significantly Transition of GH release from infancy to puberty,
compared to lean individual, but also to nonobese diabetic and into adulthood and old age
patients (172, 278, 430). This suggests that the impact of obe-
In utero and infancy
sity compounds diabetic derangements in GH release. Impor-
tantly, intensive insulin treatment to reverse hyperglycemia in In Man, fetal GH in circulation is detected by 10 weeks of
type I diabetic patients is associated with a return of circulat- gestation. Fetal GH originates independent from maternal or
ing levels of IGF-1 to that seen in nondiabetic individuals (10). placenta-derived GH, with data demonstrating an elevation
Again, this observation suggests that normalized glucose lev- of GH in fetal blood when compared to maternal blood and
els following insulin treatment may be the overriding factor amniotic fluid assessed at the same time (203). This is in
that modulates GH-axis function relative to hyperglycemia, agreement with prior observations that demonstrate fetal pitu-
and therefore insulin may serve as a critical regulator of altered itary GH synthesis (364), further demonstrating a rise in fetal
GH release in both type I and II diabetic patients, and relative GH release from 7 and 20 weeks of gestation. Collectively,
to hyperglycemia in general. This is a very different possibil- these observations suggest that maternal GH does not play a
ity from rodent studies as discussed above. significant role in the release of fetal GH. Indeed, fetal GH
is pulsatile [as verified in fetal calves and lambs (109) (31)]
and thus likely under fetal hypothalamic control. This sug-
Secretion of GH Throughout Lifespan gest that the emergence of feedback systems in utero that
occur independent of maternal systems. To this extent, fetal
As detailed above, the production and release of GH is tightly IGF-I feedback contributes to the decrease in utero GH levels
controlled by physiological needs, and in particular metabolic between week 20 of gestation and birth (203).
requirements. This is of specific importance during adult- While profound GH deficiency in utero contributes to
hood, however the biological roles of GH varies greatly with reduced longitudinal bone growth (176), and GH-gene defects
age and reproductive status, with sexual maturation and the contribute to a slight decrease in body length at birth [approx.
awakening of the reproductive axis largely determining GH 2 cm when compared to controls (121)], the overall actions
patterning and release during childhood and in puberty. This of fetal GH in gestation are not yet fully understood. Indeed,
section will document release of GH relative to sexual matu- attempts to define specific GH actions are somewhat com-
ration and ageing, focusing on childhood, puberty, adulthood, promised by the complexity of factors that contribute to fetal
and old age. Given differential effects on steroid hormones development and growth. To this extent, GH may not criti-
on GH action and release, this section will further detail sex cally contribute to normal fetal growth, and reports of nor-
specific alterations in GH output, and will briefly consider the mal fetal growth in the absence of fetal GH exist (434). Full
effects of estrogens, pregnancy, and lactation on GH output consideration of fetal-specific factors that contribute to fetal
in females. Circadian patterns of GH release in Man change growth cannot be discussed here, and the reader is advised
considerably relative to sleep and stress, and thus the effects to consult recent expert reviews on this topic (203). Diffi-
of sleep and stress on GH release will be discussed. While culties to define potential effects of GH on fetal physiology
observations are predominantly limited to Man, other species are further complicated by potential maternal GH effects on

Volume 6, April 2016 705


706
Growth Hormone Axis

Table 1 Growth Hormone Pulsatility Parameters in Humans According to Gender and Age

Gender Age Burst No. of pulses


(n) (years) Total GH Secretion rate Mean GH Burst mass amplitude Basal ApEn per 24 h Sampling frequency/period Reference

Prepubertal
M (17) 10.6 n/a 130 ug/L.24 h 2.5 ug/L∗ 6 ug/L∗ n/a 8.7 ug/L.min 0.64∗ 22 (11 × 2) 10 min, 12 h starting at 1800 h (9)
M (5) 9.5 n/a n/a 3.4 ug/L n/a n/a n/a 0.90 n/a 30 min, 24 h, starting at 1000 h (2)
F(11) 9.9 n/a 148 ug/L.24 h 2.8 ug/L∗ 8 ug/L∗ n/a 8.5 ug/L.min 0.60∗ 18 (9 × 2) 10 min, 12 h starting at 1800 h (9)
F (6) 9.1 n/a n/a 3.6 ug/L n/a n/a n/a 0.70 n/a 30 min, 24 h, starting at 1000 h (2)
Adolescent
M (13) 14.9 n/a 364 ug/L.24 h 5.9 ug/L∗ 14 ug/L∗ n/a 7.2 ug/L.min 0.61∗ 22 (11 × 2) 10 min, 12 h starting at 1800 h (9)
F (17) 14.1 n/a 270 ug/L.24 h 3.7 ug/L∗ 8 ug/L∗ n/a 8.2 ug/L.min 0.74∗ 24 (12 × 2) 10 min, 12 h starting at 1800 h (9)
Adult
M (6) 27 15 ug.24 h n/a n/a 1.8 ug/L n/a 0.9 ug/24 h 0.19 8.5 10 min, 24 h starting at 0900 h (5)
M (11) 20-28 n/a 73.3 ug/L.24 h 1.4 ug/L 3.82 ug/L n/a 3.3 ug/L.24 h 0.24 19 10 min, 24 h starting at 0800 h (3)
M (8) 26 n/a n/a 1.3 ug/L 3.6 ug/L∗ n/a 0.2 ug/L∗ 0.42 n/a 10 min, 7.5 h starting at 0630 h (6)
M (41) 43 35.8 ug/L.24 h n/a 0.4 ug/L 4.6 ug/L 4.6 ug/L 3.5 ug/L 0.34 12 10 min, 24 h (starting time not specified) (8)
F(9) 28 60 ug.24 h n/a n/a 4.9 ug/L n/a 2.3 ug/24 h 0.44 13 10 min, 24 h starting at 0900 h (5)
F (6) 22 n/a n/a 3.2 ug/L 5.1 ug/L∗ n/a 0.4 ug/L∗ 0.55 n/a 10 min, 7.5 h starting at 0630 h (6)
F (59) 38 53.1 ug/L.24 h n/a 0.5 ug/L 3.8 ug/L 3.8 ug/L 6.9 ug/L 0.43 11 10 min, 24 h (starting time not specified) (8)
Comprehensive Physiology

Volume 6, April 2016


Volume 6, April 2016
Comprehensive Physiology

Aged
M (45) 64.2 n/a n/a 0.3 ug/L n/a n/a n/a 0.60 n/a 20 min, 24 h, starting at 0600 h (1)
M (16) 70.2 n/a n/a n/a 2.9 ug/L 0.1 ug/L.min 0.8 ug/L.12 h 0.65 11.6 (5.8 × 2) 20 min, 12 h, starting at 2000 h (4)
F (38) 64.6 n/a n/a 0.5 ug/L n/a n/a n/a 0.81 n/a 20 min, 24 h, starting at 0600 h (1)
F 12) 63 n/a n/a 5.8 ug/L n/a 0.06 ug/L/h 0.57 n/a 10 min, 6 h, starting at 0800 h (7)
GHRH-R mutants
M(4) 23-30 n/a n/a 0.04 ug/L 0.05 ug/L <1% of 0.02-0.03 ug/L 1.284 15.3 10 min, 24 h starting at 0800 h (3)
normal
M (1) 24 2.7 ug.24 h n/a n/a 0.10 ug/L <4% of 0.5 ug/24 h 0.82 21 10 min, 24 h starting at 0900 h (5)
normal∗
F (1) 23 1.6 ug.24 h n/a n/a 0.06 ug/L <3% of 0.2 ug/24 h 1.17 23 10 min, 24 h starting at 0900 h (5)
normal∗

1. Charmandari E, Pincus SM, Matthews DR, Dennison E, Fall CH, and Hindmarsh PC. Joint growth hormone and cortisol spontaneous secretion is more asynchronous in older females than in
their male counterparts. J Clin Endocrinol Metab 86: 3393-3399, 2001.
2. Ilias I, Ghizzoni L, and Mastorakos G. Orderliness of cortisol, growth hormone, and leptin secretion in short-normal pre-pubertal boys and girls. Med Sci Monit 15: CR242-CR247, 2009.
3. Maheshwari HG, Pezzoli SS, Rahim A, Shalet SM, Thorner MO, and Baumann G. Pulsatile growth hormone secretion persists in genetic growth hormone-releasing hormone resistance. Am J
Physiol Endocrinol Metab 282: E943-E951, 2002.
4. Muniyappa R, Sorkin JD, Veldhuis JD, Harman SM, Munzer T, Bhasin S, and Blackman MR. Long-term testosterone supplementation augments overnight growth hormone secretion in healthy
older men. Am J Physiol Endocrinol Metab 293: E769-E775, 2007.
5. Roelfsema F, Biermasz NR, Veldman RG, Veldhuis JD, Frolich M, Stokvis-Brantsma WH, and Wit JM. GH secretion in patients with an inactivating defect of the GHRH receptor is pulsatile:
evidence for a role for non-GHRH inputs into the generation of GH pulses. J Clin Endocrinol Metab 86: 2459-2464, 2001.
6. Veldhuis JD, Farhy L, Weltman AL, Kuipers J, Weltman J, and Wideman L. Gender modulates sequential suppression and recovery of pulsatile growth hormone secretion by physiological
feedback signals in young adults. J Clin Endocrinol Metab 90: 2874-2881, 2005.
7. Veldhuis JD, Keenan DM, Bailey JN, Adeniji A, Miles JM, Paulo R, Cosma M, and Soares-Welch C. Testosterone supplementation in older men restrains insulin-like growth factor’s dose-dependent
feedback inhibition of pulsatile growth hormone secretion. J Clin Endocrinol Metab 94: 246-254, 2009.
8. Veldhuis JD, Roelfsema F, Keenan DM, and Pincus S. Gender, age, body mass index, and IGF-I individually and jointly determine distinct GH dynamics: analyses in one hundred healthy adults.
J Clin Endocrinol Metab 96: 115-121, 2011.
9. Veldhuis JD, Roemmich JN, and Rogol AD. Gender and sexual maturation-dependent contrasts in the neuroregulation of growth hormone secretion in prepubertal and late adolescent males
and females–a general clinical research center-based study. J Clin Endocrinol Metab 85: 2385-2394, 2000.

707
Growth Hormone Axis
708
Growth Hormone Axis

Table 2 Growth Hormone Pulsatility Parameters in Mammalian Species According to Gender, Hormonal Status, and Age

Species gender Age Pulsatile secretion Burst No of pulses Sampling frequency/


or GNX (n) (years) Total GH rate Mean GH Burst mass amplitude Basal ApEn per 24 h period Reference

Macaques
M (4) 5-6 y n/a n/a 2.4 ng/mL 18.5 ng/mL n/a n/a 0.50 8.5 15 min, 15 h starting (4)
(5.3 × 1.6) at 0700 h
M (4) 5-6 y n/a n/a 2.4 ng/mL 7 ng/mL 1.5 ng/mL.min n/a 0.50 11.2 15 min, 15 h starting
(7.0 × 1.6) at 0700 h
Horses Gelding 1.7 y 441 ug/L.8 h 435 ug/L.8 h 2.8 ug/L 2.8 ug/L 0.86 ug/L.min 6.5 ug/L.8 h 0.60 25.8 5 min, 8 h starting at (1)
(12) (8.6 × 3) 2200 h
Goats
M (5) 1y n/a n/a n/a 155 ng/mL 38 ng/mL <0.8 ng/mL n/a 10.7-13.4 15 min, 24 h starting (5)
(4-5 × 2.7) at 1000 h
ORCHI (11) 1-2 y n/a n/a n/a 100-155 ng/mL 40-78 ng/mL <0.8 ng/mL n/a 10.7-18.7 15 min, 6 h starting
(4-5 × 2.7) at 1000 h
OVX (6) 2-4 y n/a n/a n/a n/a n/a <0.8 ng/mL 0.65 ∼10.7 15 min, 9 h starting (10)
(∼4 × 2.7) at 0600 h
Ewes
OVX (7) 2-4 y n/a n/a n/a n/a 3.0 ug/L 0.7 ng/mL 0.93 26.4 5 min, 4 h starting at (9)
(4.4 × 6) 0900 h
OVX (5) 2-3 y n/a n/a n/a n/a 38.4 ng/mL 17 ng/mL 1,02 18.4 10 min, 6 h starting (2)
(4.6 × 4) at 0830 h
Comprehensive Physiology

Volume 6, April 2016


Rats

Volume 6, April 2016


F (5) 0.3 y n/a n/a n/a n/a 75 ng/mL 8 ng/mL 0.95 14.0 15 min, 6 h starting (6)
(3.5 × 4) at 1000 h
Comprehensive Physiology

OVX (6) 0.3 y n/a n/a n/a n/a 70 ng/mL 5 ng/mL 0.55 14.0 15 min, 6 h starting
(3.5 × 4) at 1000 h
M (8) 0.3 y n/a n/a 36 ng/mL n/a 99 ng/mL 15 ng/mL 0.79 6.1 20 min, 9 h starting (8)
(2.3 × 2.7) at 1000 h
Mice
M (7) 0.2 y 1257 ng/mL.6 h 1200 ng/mL.6 h n/a n/a 301 ng/mL.6 h 57 ng/mL.6 h 0.46 18.4 10 min, 6 h starting (7)
(4.6 × 4) at 0600 h
M (6) 0.2 y 881 ng/mL.6 h 724 ng/mL.6 h n/a n/a 301 ng/mL.6 h 87 ng/mL.6 h 0.50 18.0 10 min, 6 h starting
(4.5 × 4) at 0600 h
M (6) 0.2 y 280 ng/mL.6 h 200 ng/mL.6 h n/a n/a 40 ng/mL.6 h 80 ng/mL.6 h 0.75 22.0 10 min, 6 h starting (3)
(5.5 × 4) 1000 h
M (5) 0.7 y 280 ng/mL.6 h 200 ng/mL.6 h n/a n/a 55 ng/mL.6 h 80 ng/mL.6 h 0.50 16.0 10 min, 6 h starting
(4.0 × 4) 1000 h

1. de Graaf-Roelfsema E, Veldhuis PP, Keizer HA, van Ginneken MM, van Dam KG, Johnson ML, Barneveld A, Menheere PP, van Breda E, Wijnberg ID, and van der Kolk JH. Overtrained horses
alter their resting pulsatile growth hormone secretion. Am J Physiol Regul Integr Comp Physiol 297: R403-R411, 2009.
2. Grouselle D, Chaillou E, Caraty A, Bluet-Pajot MT, Zizzari P, Tillet Y, and Epelbaum J. Pulsatile cerebrospinal fluid and plasma ghrelin in relation to growth hormone secretion and food intake
in the sheep. J Neuroendocrinol 20: 1138-1146, 2008.
3. Hassouna R, Zizzari P, Tomasetto C, Veldhuis JD, Fiquet O, Labarthe A, Cognet J, Steyn F, Chen C, Epelbaum J, and Tolle V. An early reduction in GH peak amplitude in preproghrelin-deficient
male mice has a minor impact on linear growth. Endocrinology 155: 3561-3571, 2014.
4. Lado-Abeal J, Veldhuis JD, and Norman RL. Glucose relays information regarding nutritional status to the neural circuits that control the somatotropic, corticotropic, and gonadotropic axes in
adult male rhesus macaques. Endocrinology 143: 403-410, 2002.
5. Mogi K, Li JY, Suzuki M, Sawasaki T, Takahashi M, and Nishihara M. Characterization of GH pulsatility in male Shiba goats: effects of postpubertal castration and KP102. Endocr J 49:
145-151, 2002.
6. Painson JC, Veldhuis JD, and Tannenbaum GS. Single exposure to testosterone in adulthood rapidly induces regularity in the growth hormone release process. Am J Physiol Endocrinol Metab
278: E933-E940, 2000.
7. Steyn FJ, Huang L, Ngo ST, Leong JW, Tan HY, Xie TY, Parlow AF, Veldhuis JD, Waters MJ, and Chen C. Development of a method for the determination of pulsatile growth hormone secretion
in mice. Endocrinology 152: 3165-3171, 2011.
8. Tolle V, Bassant M-H, Zizzari P, Poindessous-Jazat F, Tomasetto C, Epelbaum J, and Bluet-Pajot M-T. Ultradian rhythmicity of ghrelin secretion in relation with GH, feeding behavior, and
sleep-wake patterns in rats. Endocrinology 143: 1353-1361, 2002.
9. Veldhuis JD, Fletcher TP, Gatford KL, Egan AR, and Clarke IJ. Hypophyseal-portal somatostatin (SRIH) and jugular venous growth hormone secretion in the conscious unrestrained ewe.
Neuroendocrinology 75: 83-91, 2002.
10. Yonezawa T, Mogi K, Li JY, Sako R, Manabe N, Yamanouchi K, and Nishihara M. Negative correlation between neuropeptide Y profile in the cerebrospinal fluid and growth hormone pulses
in the peripheral circulation in goats. Neuroendocrinology 91: 308-317, 2010.

709
Growth Hormone Axis
Growth Hormone Axis Comprehensive Physiology

fetal growth. Indeed, maternal GH modulates placental func- is associated with the greatest period of change in GH levels
tion, enhancing fetal growth by enhancing nutrient exchange and patterning that is observed throughout life. Emerging
between the mother and fetus (168). Mechanisms of maternal- feedback from sex-steroid hormones is largely responsible
enhanced fetal growth will not be discussed here, as these for the transition of altered GH release throughout puberty,
effects largely diverge from the neuroendocrine regulation as demonstrated following administration of estrogen or
of fetal GH release (see below maternal control of GH in testosterone to individuals with ovarian (330) or testicular
pregnancy). (173, 303, 537) dysfunction that results in impaired steroid
Following birth, growth continues at the same rate as is hormone production. In all instances, steroid hormone treat-
seen in utero until approximately 6 to 10 months of age, ments mostly improve GH output to that normally seen in
after which growth slows during childhood. The appearance healthy individuals. Regarding GH release, pubertal matura-
of GH, and onset of pulsatile release occurs within the first tion is associated with a dramatic rise in GH output. The first
hours of birth. This is characterized by the emergence of GH comprehensive study to document this described progressive
secretory bursts within the first day/night (120). The amount changes in GH output in boys and girls, reflecting an increase
of GH produced relative to the surface area of the newborn in mean GH levels that occur largely in response to an increase
greatly exceeds that seen in childhood or adulthood. Prema- in GH pulse amplitude, and not GH pulse frequency (441).
ture birth is associated with increased GH secretion, thought Increased GH output reflected progression through pubertal
to occur as a consequence of withdrawal of feedback by IGF-1 maturation, as assessed through breast development in girls
(174). and testicular volume in boys. In girls, mean GH output
correlated negatively with BMI, whereas this was not the case
for boys. These observations were verified following the lon-
Childhood
gitudinal assessment of GH output in developing boys, adding
Estimates of GH secretion rates during childhood (prior to that alterations in GH output relative to pubertal maturation
puberty) range between 100 and 600 μg/day (228, 324, 325, does not occur in response to altered GH clearance/half-life
441, 442, 552). Given variable observations between studies, (325).
accurate estimates of GH release throughout childhood may The mechanism of increased GH release throughout
be best-inferred following longitudinal assessment. Longitu- puberty is unique, in that the rise in GH secretion at this time
dinal assessment of GH output in healthy boys ranging from occurs alongside an elevation in circulating levels of IGF-1
9.5 years of age to close to 12 years of age revealed a fourfold (hypersomatotropism) (441). In this regard, anticipated feed-
range in GH output parameters between individuals, whereas back between GH and IGF-1 does not predict GH release.
within individual variability was minimal (325). Within this Moreover, this is not coupled with hepatic GH resistance,
cohort, GH pulse amplitude largely contributed to between- as is often seen in conditions wherein GH/IGF-1 balance is
individual variability in mean GH release. By comparison, disrupted (174). It is thought that alterations in the sensitivity
pulse frequency was mostly conserved. No change in GH of hypothalamic-pituitary feedback mechanisms may account
release (within individuals) was observed with advanced age, for derangements in GH/IGF-1 interactions during puberty [as
and thus it is inferred that GH release in childhood remains discussed in (174)]; however, this remains to be confirmed.
stable. This observation matches that from earlier reports, con- Despite continued production of adult sex-steroid hormones,
firming little to no change in mean GH concentrations, mean the duration of this period of GH/IGF-1 hypersecretion is lim-
day-time and night-time GH concentrations, mean night-time ited, and it therefore remains unclear why GH output declines
GH pulse amplitude and the number of GH secretory events in to prepubertal and below prepubertal levels with increased
boys aged under 9 years of age when compared to prepubertal age. Of interest, feedback mechanisms associated with the
boys over 9 years of age (441). By contrast, all parameters in accumulation of adipose tissue may contribute to the progres-
prepubertal girls at 8 years of age increased when compared sive decline in GH release following pubertal maturation, as
to girls that were younger than 8 years of age, demonstrating was demonstrated in early-adult mice in response to high fat
sex differences in the timing of the increase in GH release rel- feeding (224).
ative to pubertal maturation (441). Of interest, the differential
change in the onset of increased GH output between boys and
Adulthood
girls reflect altered growth rates documented between sexes,
wherein increased growth rate occurs earlier in puberty in An exponential decline in mean 24-h GH concentrations is
girls (158, 517). observed in men and women, starting from 18 to 25 years
of age, with GH output halving on average every 7 years
(174). This occurs regardless of sustained adult levels of
Puberty
sex hormones, and is accompanied by the gradual decline
The transition of childhood into puberty occurs alongside in circulating levels of IGF-1. A number of factors may con-
a number of critical physiological changes, mediated by tribute to this progressive decline in GH release (often referred
endocrine responses to sexual maturation of the gonads, and to as somatopause), including age, body composition (and

710 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

notably adiposity), and exercise capacity. These factors pri- will be briefly discussed below, focusing in particular on the
marily impact GH burst mass, and not GH pulse frequency role of estrogen in the regulation of GH release.
(233,548,570), suggesting that factors that impact GH release
with age act predominantly on the pituitary gland, modifying
Estrogen
pituitary output or responses to hypothalamic cues. Of inter-
est, gender determines the severity of the impact of age, obe- Pubertal maturation and the emergence of the ovulatory cycle
sity and exercise on GH output (570). Compared to men, is associated with an increase in serum GH levels, and the
healthy premenopausal females are protected from the impact emergence of alterations in GH output that are correlated
of age and relative adiposity on daily GH output, whereas with changes in serum estradiol levels. Assessment of pul-
this effect is lost in postmenopausal females in the absence of satile GH output in pubertal girls revealed a twofold to three-
gonadal hormone replacement therapy (102). While the clear- fold increase in peak GH release from the onset of puberty to
ance of GH secretion between males and females is conserved, menarche. This increase is correlated with rising circulating
GH output in females is greatly amplified, and characterized levels of estrogen (571), suggesting that endogenous estradiol
by approximately twofold greater peaks in GH release (541). production in females further increases GH secretion. Indeed,
Clinical observations [as detailed previously (551)] suggest initial observations suggested a twofold rise in GH secretion
that an elevation in endogenous estrogen in females con- in women across the menstrual cycle (151). This was con-
tributes to increased peak GH output through; (i) accentuating firmed following the assessment of pulsatile GH release in
feedback by GH following a GH secretory event (545), (ii) females, wherein a twofold rise in GH release was observed
by augmenting somatostatin withdrawal-induced GH release toward the late follicular stage of the menstrual cycle (140).
(545), (iii) by promoting endogenous actions of GHRH (390) To this extent, use of estrogens for superovulation induces
and the potency of exogenous GHRH (547) on GH release, a significant rise in GH release (579). It is now well estab-
(iv) by reducing pituitary responsiveness to somatostatin (59), lished that circulating patterns of GH release in males versus
and (v) by opposing GH feedback on GHRH stimulation (11). females vary considerably, with increased peak GH output
Given the close relationship between GH output and steroid in females occurring as a consequence of increased levels of
hormones (and in particular estrogen), it comes as no surprise circulating levels of estradiol. To this extent, treatment of men
that GH release within females varies considerably relative to with diethylstilbestrol (a potent estrogen) results a shift in the
reproductive status. pulsatile pattern of GH release to reflect that commonly seen
in females (151).
Alterations in GH output in response to estrogens is
thought to occur as a consequence of altered negative feed-
Divergent control of GH release in females relative back, wherein the actions of IGF-1 on hypothalamic com-
to reproductive status ponents of the GH-axis are greatly diminished (574). This is
As detailed earlier, a number of factors contribute to alter GH somewhat controversial (35), with evidence suggesting a com-
output between females and males following sexual matura- plex relationship between IGF-1, estrogen, and GH output.
tion, and throughout life (Fig. 7). Importantly, sexual differ- At low dosages, estrogens increase GH output, whereas high
entiation may occur prior to birth, with the prenatal steroid dosages of estrogens may decrease levels of bioactive IGF-1.
hormone environment regulating critical differences in the Moreover, the route of estrogen administration significantly
number of GHRH neurons, their responses to postpubertal alters the effect of estradiol on GH release. Assessment of
steroids, adult hypothalamic synaptic organization (including pulsatile GH output in pre- and postmenopausal women fol-
synaptic inputs and glial cell morphology), and the number lowing oral versus transdermal estradiol treatment revealed
and responsiveness of somatotroph that dictate adult GH out- divergent effects on GH release. In two independent stud-
put. This article will focus predominantly on the differen- ies, oral estradiol treatment increased 24 h mean and pul-
tiation of GH secretory patterns that occur during pubertal satile GH release and decreased circulating levels of IGF-1,
maturation, and in adulthood. The reader is asked to refer to whereas transdermal estrogen treatment increased circulat-
excellent reviews on neonatal development for a more com- ing levels of IGF-1 in postmenopausal to that seen in pre-
prehensive overview of factors that predict GH release prior menopausal women without altering the pulsatile release of
to birth (92). Moreover, while differences in GH release most GH (36, 569). Thus, it is thought that oral administration of
likely occur as a consequence of steroid hormone feedback, estrogen increases GH release through lowering circulating
further changes in GH output are observed specific to dif- levels of IGF-1, thereby reducing negative feedback. These
ferent metabolic states that occur as a consequence of addi- observations further highlight that the reduced GH output fol-
tional reproductive pressures that are encountered throughout lowing menopause is not due to a reduction in circulating
female adult life. Of these, altered GH output relative to the levels estrogen (569). To this extent, neither oral nor transder-
menstrual cycle, and pregnancy and lactation demonstrate mal estrogen treatment in postmenopausal women can reverse
additional feedback mechanisms between central and periph- age-related reductions in spontaneous or GHRH-stimulated
eral regulators of GH release that are not seen in males. These GH and IGF-I release (36).

Volume 6, April 2016 711


Growth Hormone Axis Comprehensive Physiology

Figure 7 Differential GH secretory patterns between males and females. Mechanisms that promote GH output in adulthood are established during
pubertal maturation. For males, this results in the establishment of adult-specific GH release by 18 to 25 years of age, and the progressive decline
in peak and total GH release with age. Differential patterns and amounts of GH release between males and females occur as a consequence of
altered feedback to the hypothalamus and anterior pituitary gland, predominantly in response to circulating levels of estrogen. Increased circulating
levels of estrogen are associated with an overall rise in peak amplitude, pulse frequency, and total GH release in females when compared to males.
A further rise in peak and total GH release in females is observed during the follicular stage of the ovarian cycle, the period characterized by a rise
in circulating levels of estrogen (peaking prior to ovulation). Feedback from placental-derived GH to the anterior pituitary gland contributes to the
suppression of maternal GH release during pregnancy. Resumption of maternal GH output following birth is critical for sustained milk production
through prevention of involution of mammary gland tissue. Acting through direct mechanisms, or indirectly through insulin-like growth factor-1
(IGF-1), GH is thought to further promote the transport of nutrients to maternal milk.

712 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

Pregnancy (535). Comprehensive reviews should be consulted for addi-


tional information specific to maternal fetal GH interactions,
Initial observations suggested a progressive rise in maternal
and the role of placental GH on fetal growth (177).
GH (268) and IGF-I (190) in pregnancy; however, subsequent
use of selective antibodies to distinguish maternal versus pla-
cental GH demonstrated the replacement of maternal GH by Lactation
placental GH relative to placental development. Specifically, a
The withdrawal of the placenta, and therefore placental GH,
progressive rise in circulating levels of placental-derived GH,
is associated with the resumption of maternal GH production.
starting from 20 weeks of gestation was observed (134, 150).
Observations from all mammalian species assessed to date
More recent direct measures confirmed the progressive rise
argue for a critical role for GH in regulating milk production
of placental GH starting from 8 to 10 weeks of pregnancy,
and release. Indeed, GH treatment enhances milk produc-
peaking at 35 to 36 weeks of pregnancy (309). This progres-
tion in humans (346), and GH treatment may therapeutically
sive rise in placental GH replaces maternal GH, with maternal
improve lactational insufficiency in mothers with preterm
levels gradually decreasing to near undetectable amounts in
infants (184). In dairy ruminants, GH treatment improves
circulation (134,150). In this regard, it was proposed that pla-
milk yield (32) and milk triglyceride content (310). In lactat-
cental GH or others pregnancy-related factors (placental lac-
ing rats the inhibition of GH release causes a reduction in milk
togen, IGF_1) act at the level of the anterior pituitary gland to
yield and milk fat (144). IGF-1 is positively related to milk
suppress GH release (122). However comprehensive data to
fat composition and milk yield (186), and thus it is thought
exclude potential direct inhibitory effects of placental GH on
that GH induces IGF-1 release to regulate milk composition
the human hypothalamus do not exist. Unlike maternal GH,
(143). Of interest, the reintroduction of previously removed
placental GH release is not pulsatile (134), and thus contin-
pups and the resumption of the suckling reflex induces a surge
ued feedback should completely silence maternal GH release.
in maternal plasma GH release (435). As the suckling reflex
Placental GH is secreted solely into maternal circulation, and
is accompanied by a concomitant increase in circulating GH
thus the function of placental GH is not completely clear.
and prolactin release (435, 456), common mechanisms may
Given the actions of GH in regulating energy metabolism,
modulate prolactin and GH release. Alternatively, suckling-
prolonged elevation of placental GH may induce relative
induced changes in prolactin release may facilitate increased
insulin resistance, allowing for the preferential transfer of glu-
GH output. Over the course of development, lactotrophs dis-
cose to the fetus, while shifting maternal energy homeostasis
play transient coexpression of GH secretory capability (285)
to encourage maternal dependence on lipolysis for energy
and both systems are under common control of endogenous
(177). This remains to be verified, especially as pregnancy
opiates (435). These observations suggest close integration
in the mother (although highly variable between individu-
of the mechanisms that facilitate production and release of
als) is commonly associated with weight gain due to increase
GH and prolactin. In the context of lactation, both GH and
adiposity (http://www.ncbi.nlm.nih.gov/books/NBK32815/),
prolactin are required to maximize milk yield, milk fat con-
and thus a lipolytic state normally associated with reduced
tent and the inhibition of mammary gland involution (143),
GH secretion.
and thus prolactin and GH may act synergistically to modulate
Maternal GH deficiency documented prior to pregnancy
lipolytic function of mammary tissue, as suggested previously
does not impact birth outcomes (111), suggesting that prevail-
(25, 26).
ing maternal GH levels does not impair fetal development. By
IGF-1 is a survival factor in mammary tissue morphology,
contrast, intrauterine growth retardation is associated with low
mediating mammogenesis (143) and lactogenesis (186). GH
or deficient maternal IGF-1 and placental GH levels (350).
acts directly on the mammary epithelium to induce paracrine
This suggests that maternal GH deficiency specific to preg-
IGF-1 production by the mammary stroma (143). Accord-
nancy may compromise fetal growth. Fetal production of GH
ingly, GH may modulate established effects of IGF-1 as a
occurs by 12 weeks of gestation (34), whereas placental GH
survival factor in mammary tissue. Should this occur, lacta-
does not cross the placenta (150). Therefore, it is unlikely
tion must be associated with a rise in GH release. Morpho-
that maternal GH directly contributes to fetal growth. Rather,
logical changes within pituitary gland contradict this notion,
maternal-derived GH may be critical for the exchange of fac-
as structural changes within the anterior pituitary gland favor
tors essential for optimal fetal growth (202). Given limited
the production of lactogens. Gestation and lactation in humans
correlative evidence for the role of placental and maternal
(482) and rats (187) is associated with pituitary hypertrophy
IGF-1 in fetal growth, it is thought that these maternally
and hyperplasia. In lactating rats, this is characterized by the
derived hormones likely contribute to maternal responses to
presence of numerous hypertrophied lactotrophs, a significant
pregnancy, potentially mediating maternal nutrient transfer.
increase in the percentage of lactotrophs, and a corresponding
Indeed, treatment of pregnant pigs with GH during early preg-
decrease in somatotroph number. This morphological change
nancy removes maternal constraints on fetal growth, resulting
is reversed following weaning, and therefore the cessation
in increased size of prodigy at birth (167). Recent observations
of suckling (187). In humans, lactation is known to reduce
confirmed that these positive effects occurred predominantly
GH mRNA expression and increase prolactin mRNA expres-
in response to enhanced maternal nutrient transfer to the fetus
sion (482). Moreover, as lactation proceeds, somatotroph cells

Volume 6, April 2016 713


Growth Hormone Axis Comprehensive Physiology

transdifferentiate into mammosomatotrophs (131) with the sleep deprivation (553). Although there is a strong link
ability to secrete prolactin and GH. These transdifferentiated between sleep-wake cycle and GH secretion, it is difficult
cells eventually develop into lactotrophs (554). Therefore, due to define whether sleep modulates GH secretion or whether
to the extent of observed transdifferentiation coupled with GH influences sleep-wake cycles. SWS seems to precede the
morphological changes within the gland, one may anticipate nocturnal GH burst but some studies observed that the noc-
an overall reduction in GH output (131). This contradicts turnal GH burst precedes the beginning of the SWS or that
anticipated actions of GH on IGF-1, and consequential GH a third of SWS episodes are not associated to GH secretion
and IGF-1 mediated effects on maternal milk quality and (539).
mammary gland morphology. At any rate, the transdifferen- Recent reviews have described the effects of GHRH and
tiation of mammosomatotrophs remains an interesting obser- GH secretagogues on sleep-wake cycles (260, 519). In rats,
vation that needs to be further validated with modern genetic rabbits and humans, exogenous GHRH promotes nonrapid
methods for tracing cell lineages. Given potential morpho- eye movement (NREM) sleep and suppression of endoge-
logical changes within the pituitary gland to favor lactogen nous GHRH (with antagonists, antibodies, somatostatin stim-
output, should an increase in GH release be observed in lac- ulation or high doses of GH or IGF-1 through negative feed-
tation, this shift in GH output may occur as a direct con- back) inhibits NREM sleep (386). Indeed, GHRH increases
sequence of altered hypothalamic control or altered soma- the duration and/or intensity of NREM sleep, including SWS,
totrophs responses to hypothalamic (and possibly peripheral) with a greater efficacy of episodic than continuous GHRH
feedback. Alternatively, as prolactin may increase circulating administration (323). After sleep deprivation, GHRH also
levels of IGF-1, acting directly within the liver (368), the role enhances NREM sleep (463). The sleep-promoting effect of
of GH as a key regulator of IGF-1 in lactation may be dimin- GHRH is observed after ip and icv injections as well as after
ished, with prolactin directly promoting the anti-involution direct injections in the preoptic area in male rats (385, 596)
and lipolytic actions of IGF-1. However, this seems unlikely, and in healthy young men (261, 323, 410, 484). Surprisingly,
as serum IGF-1 levels do not appear to contribute to the mam- an opposite effect is reported in young women (329). In trans-
motrophic actions of PRL (132). Given the well-established genic mice with reduced GHRH, NREM sleep is suppressed
role of GH in sustaining optimal lactation, it is surprising during both the light dark periods but REM sleep is unmodi-
that mechanisms that define GH release in lactation remains fied (595).
completely unexplored. Genetic alterations that compromise GHRH transmission
are associated with reduced NREM sleep in both rodents
and in humans (260). In dwarf mice with transgenic over-
GH regulation and sleep expression of human GH to sites of tyrosine-hydroxylase
Bidirectional interactions between the activity of the GH/IGF- synthesis in the brain, plasma endogenous GH, IGF-1, and
1 axis and sleep regulation have been observed (210, 483, GHRH are nearly undetectable and the duration of NREM
540). In humans, GH secretion is directly linked to the sleep- sleep and total sleep time are reduced while wakefulness is
wake cycle. Indeed, GH is secreted preferentially during sleep increased (595). Similarly, the lit/lit mouse with nonfunc-
and maximal GH secretory burst occurs within minutes after tional GHRH receptors displays reduced NREM sleep (and
the onset of slow-wave sleep (SWS). When the sleep cycle slightly decreased REM sleep as well) during the light period
is delayed or advanced, GH secretion is either delayed or and sc GHRH infusion restores NREM sleep values similar to
advanced to coincide with the first episode of sleep. In rodents, those observed in wild-type mice (383). In spontaneous dwarf
the link between GH secretion and sleep-wake cycles are more rats with a recessive abnormality that decreases GHRH-R and
difficult to confirm, as sleep patterns in rodents are defined by GH levels (dw/dw), reduced NREM sleep during the light and
multiple and sporadic wake/sleep phases over a 24 h period dark cycles and reduced REM sleep during the light period is
(529). observed (384). In contrast, transgenic mice over-expressing
The association between GH secretion and sleep is con- GH show increased NREM (and REM) sleep during the light
served in subjects with sleep disorders, wherein altered period (188).
episodes of wakefulness and sleep are observed throughout Growth hormone secretagogues (GHS) and ghrelin are
the day; the beginning of the SWS precedes the GH burst also modulators of the sleep-wake cycle. In healthy young
(423). In healthy subjects, when sleep is delayed by several men, 1-week oral treatment with the GHS MK-0677 increased
hours, the nocturnal GH burst is also delayed (179, 427, 567). SWS and decreased wakefulness (103). In elderly subjects,
In narcoleptic subjects, the nocturnal GH secretory burst is GHS does not appear to affect the sleep-promoting effects of
considerably reduced or absent (44, 94, 200). Sleep depriva- GHRH (103). When GHRP-6, another GHS, is administered
tion also impacts GH secretion, and the nocturnal GH secre- intravenously in a pulsatile manner (four injections during
tory burst is suppressed after 24 h of sleep deprivation. The the night), NREM sleep stage 2 increases as well as nocturnal
absence of peak nocturnal GH secretion is compensated for GH secretion (157). In another study by the same group,
by larger peaks during the day, whereas total GH secretion after pulsatile intranasal, oral and sublingual administration
per 24 h remains constant (58). In addition, an increase in GH of the same GHS, the effect on sleep architecture was not
secretion is observed during the early sleep cycle following as pronounced, suggesting that the observed effects depend

714 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

on the route of administration (156). Furthermore, the mode GH expression in the pups pituitary is reduced. In addition,
of administration (pulsatile versus single injection) could be maternal malnutrition induced by stress is associated with a
an important determinant of the effects of GHS on sleep, reduction in serum IGF-1 levels in the dams and the pups
as a single injection of GHRP-2 during the third period of (165).
REM sleep does not affect sleep (355). Hexarelin, in contrast, The physiological effects of stress can be mediated
decreased the amount of SWS and increased GH secretion through many different mechanisms, including increased
(155). Possible negative feedback of GH on GHRH may have serum concentrations of proinflammatory cytokines and glu-
induced a reduction in SWS in this latter study. cocorticoids (GCs), as well as impaired actions of the GH-
Ghrelin, after four repeated injections every hour at IGF-1 axis. Neuroendocrine responses to stress involve the
the beginning of the dark cycle, increases NREM sleep activation of the HPA axis with increase in hypothalamic CRH
and decrease REM sleep in young men, but not young mRNA and serum GCs level. Data concerning the actions of
women, when administered alone or coadministered with GCs on GH secretion are contradictory: GCs increasing or
GHRH (270) (271,272), reaffirming potential sex-differences reducing GH secretion depending on the experimental con-
between sleep and GH release. Ghrelin also increases SWS ditions. Dexamethasone stimulates GH release from soma-
and decreases REM sleep in elderly men but not in elderly totroph cells (392) and increases the sensitivity to GHRH in
women, although it triggers the same GH effect in both rats (371,538) and humans (370). In vivo, acute administration
sexes, indicating sexual dimorphism in the sleep promoting of dexamethasone increases the amplitude of GH secretory
but not GH-releasing effects of ghrelin (269). However, four peaks (562) and potentiates GH response to GHRH (562).
repeated injections of ghrelin in the early morning increase Some studies also reported elevated IGF-1 levels in subjects
GH secretion without affecting sleep (271,272). As with GH, exposed to dexamethasone on the short term (341, 419). In
an increase in endogenous ghrelin secretion is observed before contrast, GCs administered on the long term or with supra-
sleep onset in men but not in women (462). In young adult physiological doses inhibit growth and GH secretion in the
male rats, repeated administration of ghrelin at 3- to 4-h inter- rat (387). Direct GCs stimulatory effects in the pituitary gland
vals (one during lights-on and two during lights-off periods) are mediated through an increase in the transcriptional activity
decreased REM sleep duration (529). In ghrl−/− mice, basic of the somatotrophs (524) or by a higher affinity or receptor
sleep-wake regulatory mechanisms are not impaired (499) density for GHRH on the cell (464, 465) associated with a
but restricted feeding-induced arousal is attenuated (135). decrease in the number of somatostatin receptors but not their
Altogether, interaction between endogenous ghrelin and sleep affinity (461). At the hypothalamic level, GCs stimulatory
appear to be gender and species dependent and still needs to effects may be the consequence of altered synthesis or release
be clarified. of somatostatin and a reduction of the negative feedback from
In GH-deficient (GHD) patients, sleep-wake cycles IGF-1 on GH secretion (523). In contrast, GCs inhibitory
are characterized only in a few controversial studies effects seem to be the consequence of an increase in the
(21, 235, 359, 382, 408, 460). These studies are often difficult somatostatin tone. Indeed, inhibition of GHRH-induced GH
to interpret due to the small number of subjects, the lack of release by high doses of dexamethasone is totally absent after
proper controls or GHD of different origin (pituitary versus antisomatostatin pretreatment (565). In patients with Cush-
hypothalamic). In a placebo-controlled study, 4 months of GH ing syndrome, a decrease in GH secretion is observed (490),
therapy seem to partly reverse sleep disturbances previously accompanied by growth retardation in children (491). Chronic
observed in untreated patients (359). GHRH induced NREM stress that impairs hippocampal function in rats leads to a
sleep is a hypothalamic mechanism involving GABAergic reduction in hippocampal GH levels and restoration of hip-
neurons in the medial preoptic area and neither GH not pocampal GH by viral gene transfer reverses stress-related
IGF-1 are involved (386). Thus, as GH stimulates REM alterations (544).
sleep in both rodents and humans (20, 335) while GHRH Ghrelin is involved in both neuroendocrine and behavioral
rather promotes NREM sleep, excess of high-intensity SWS responses to stress through activation of the HPA axis (includ-
associated with decreased REM sleep in some untreated ing increases in hypothalamic CRH mRNA and serum cor-
GH-deficient subjects of pituitary origins (235) has been ticosterone). However some of the effects of ghrelin appear
interpreted as an upregulated hypothalamic GHRH release independent of the HPA axis; rats repeatedly exposed to a
secondary to the lack of negative GH feedback (104). In stressor display, a model of posttraumatic stress disorder,
GHD with hypothalamic disorders, defect in both GHRH and heighten fear learning following auditory Pavlovian fear con-
GH are associated with both reduced NREM and REM sleep ditioning through ghrelin stimulation of GH receptor in the
(519). amygdala, independently of the HPA axis (339). In humans,
however, posttraumatic stress is associated with a reduction
in nocturnal GH secretion, possibly secondary to sleep frag-
GH regulation and stress mentation in response to stress (543). Behavioral responses
Impaired bone growth is observed in children exposed to to stress may preclude anxiodepressive disorders and ghrelin
stress (457). In animal models, maternal immobilization stress has been shown to mediate metabolic-induced anxiety and
during lactation results in low body weight in the offspring. depression (282).

Volume 6, April 2016 715


Growth Hormone Axis Comprehensive Physiology

Old age and somatopause GH Axis and Longevity


This section will briefly discuss considerations pertinent to
Growth Hormone and IGF-1 levels decline during advanc-
the role of GH in ageing. For a more comprehensive overview
ing years of life. These changes (somatopause) are associ-
of the effects of ageing on GH output, the role of GH
ated with loss of vitality, muscle mass, physical function,
on accelerated ageing, and therapeutic strategies, and side
together with the occurrence of frailty, central adiposity, car-
effects of GH treatment, please consult recent comprehen-
diovascular complications, and deterioration of mental func-
sive reviews (110, 170, 557). The progressive decline of GH
tion. This phenomenon has led to recombinant human GH
release with age (16, 108, 363) culminates in levels of GH
being widely promoted and abused as an antiageing drug,
resembling that seen in adult GHD (240), and corresponds to
despite lack of evidence of efficacy and possible deleterious
decreased lean body mass, increased body fat mass, reduced
effects as indicated in the consensus statement by the Growth
bone mass, and reduced aerobic capacity. This may contribute
Hormone Research Society on the GH/IGF-I Axis in Extend-
to the development of a number of pathologies, including
ing Health Span (527). By contrast, evolutionarily conserved
increased frailty and frequency of fractures, muscle atrophy,
mutations that decrease the tone of the GH/IGF-1 axis are
and obesity (240). Despite considerable evidence from ani-
associated with extended longevity from worms to mice with
mal studies suggesting that increased GH release may accel-
some homologies extending to unicellular yeast (403). How-
erate ageing (28-30, 217), it is thought that reduced GH
ever, while mutations in IGF-1-Insulin-like receptor (Daf-
output, and an acceleration in reduced GH output with age
2) often result in a twofold to threefold lifespan extension
might accelerate the ageing process. This has spurred a num-
in Caenorhabditis elegans, corresponding mutations in mice
ber of trials wherein GH administration was considered as
are much less efficient. Such mutations and genetic models
an effective mean for slowing the ageing process [includ-
affecting the GH axis are listed on Table 3, according to their
ing (149, 211, 286, 402, 447)]. While generally promoting an
primary effect with their human counterparts.
increase in lean muscle mass at the expense of adipose tis-
Natural mutations in two genes involved in pituitary devel-
sue (149,211,286,402,447), studies often report adverse side
opment (Pou1f1, the gene encoding for the pituitary transcrip-
effects including edema (149, 211, 286, 402), fluid retention
tion factor Pit-1 in Snell mice, and Prop1, a homeobox protein
(402), hypertension (447), carpal tunnel syndrome (211,286),
involved in the transcriptional regulation of Pit-1 in Ames
and to some extent, hyperglycemia (149, 447). The efficacy
mice) are the two most potent mutations in terms of dwarfism
and risks associated with GH treatment relative to age is still
and life span extension. On both strains of mice insulin sen-
under debate, and considerable attention needs to be directed
sitivity is increased despite increased body fat. Tumor inci-
toward the mode and dosage of GH or GH secretagogues
dence is decreased and oxidative stress resistance increased.
delivery.
A homozygous PROP1 mutation has been described in 25
While still under investigation, proposed mechanisms for
patients with dwarfism originating from the Island of Krk in
impaired GH output in old-age mostly converge at the level of
Croatia. These short-statured individuals (106-152 cm) are
the hypothalamus, and it is believed that reduced GH output is
obese but they do not develop type 2 diabetes with increasing
largely due to enhanced somatostatin release, and diminished
age and do not seem to die prematurely (280).
GHRH secretion (372). In men, treatment of pyridostigmine,
A third natural mutation, a single amino acid substi-
to withdraw somatostatin and therefore to enhance GHRH
tution in the extracellular domain of the GHRH receptor,
release, recovers peripheral GH output. This results in the
induces GHRH resistance and life span extension in little
near normalization of mean 24-h serum GH and IGF-1 con-
dwarf mice. As the Snell and Ames mice, the little mice phe-
centrations in older men, when compared to young men (97).
notype includes higher insulin sensitivity despite increased
Regarding GHRH deficiency, measures are somewhat con-
body fat, lower tumor incidence, and increased stress resis-
tradicting. For example, pulsatile-GHRH infusion over three
tance. Two cohorts of patients with homozygous mutations
consecutive days failed to enhance GH output in older men
in the GHRH receptor have been reported, originating from
(173). Similarly, daily treatment of GHRH alone, or in com-
Pakistan (317) or Brazil (3, 451) with very similar heights
bination with L-arginine (to suppress endogenous somato-
(105-135 cm) and mild obesity. A reduced mean life span
statin tone), failed to normalize GH output in elderly patients
compared with the unaffected population was reported for the
(169). In this regard, impaired GH release in old age may
later (48 vs. 63 years) but no data is yet available for the for-
occur as a further consequence of impaired GHRH action.
mer. Reduced longevity relative to that of unaffected brothers
These observations stem from initial investigations that con-
and sisters (males, 56 vs. 75 year; females, 46 vs. 80 year)
sidered the effects of ageing on GH output in dogs (77-79),
has also been reported in untreated patients with isolated GH
wherein the combined roles of somatostatin and GHRH were
deficiency linked to a 6.7 kb deletion in the GH1 gene (45).
well defined. In Man, there is evidence of an age-dependent
Major GH deficiency is also observed in the Ghrh knock-
decline in circulating acyl-ghrelin levels, with ghrelin/GH
out mice on a mixed genetic background with remarkably
association more than threefold lower in the 62- to 74-year-
increased longevity accompanied by major shifts in the
old group compared with the young 18- to 28-year-old adults
expression of genes related to xenobiotic detoxification, stress
(376).
resistance, and insulin signaling (493).

716 Volume 6, April 2016


Volume 6, April 2016
Comprehensive Physiology

Table 3 Effects of Genetic Alterations in the GH/IGF-1 Axis on Longevity in Human Diseases or Genetic Variants and Mouse Models

Human diseases Lifespan Lifespan (days)


or genetic Size (% of changes Genetic Body Insulin Tumour Stress
variants Mouse models control) (%) Mutant Control background fat sensitivity incidence resistance

Pituitary Krk cohort Snella 25-33 F + 42 F 1148 ± 39 F 811 ± 80 DWC3F1 ⇑ ⇑ ⇓ ⇑


developmental M + 26 M 1037 ± 33 M 822 ± 34
defect
Amesb 33-50 F + 68 F 1206 ± 32 F 718 ± 45 heterogenous ⇑ ⇑ ⇓ ⇑
M + 49 M 1076 ± 56 M 723 ± 54
GH deficiency Itabaianinha lit/litc 50-67 F + 25 F 1070 ± 127 F 857 ± 169 C57BL/6J ⇑ nd ⇓ ⇑
Sindh M + 23 M 1093 ± 186 M 886 ± 148
Sammaun
cohorts Ghrh−/−d 55-60 F + 43 F 956 ±? F 666 ± ? C57BL/6J x ⇑ ⇑ nd ⇑
M + 51 M 928 ±? M 614 ± ? 129SV/E
GH resistance Laron syndrome Ghr−/−e 50 F + 21 F 921 ± 41 F 759 ± 41 Ola-BALB/cj ⇑ ⇑ ⇓ ⇑
M + 40 M 917 ± 55 M 656 ± 67
GH antagonism GHAf 70 F+9 F 839 ± 25 F 771 ± 26 C57BL/6J ⇑ ⇑ nd
M+4 M 790 ± 41 M 758 ± 40
GH excess Acromegaly bGH transgeneg 150 F? F? F? C57BL/6J ⇓ ⇓ ⇑ nd
M - 45 M 425 ± 22 M 773 ± ?
Decreased IGF-1 Pappa−/−h 40 F + 41 F&M F & M 698 ± 23 C57BL/6J x nd = ⇓ nd
availability in M+3 960 ± 28 F 672 ± ? 129SV/E
pericellular F + 33 F 882 ±? M 679 ± ?
environment M + 20 M 812 ± ?
Decreased LID−/−i 90 F+4 F 1087 ± 38 F 1043 ± 30 C57BL/6J ⇑ ⇓ ⇓ nd
circulating M - 18 M 836 ± 50 M 1014 ± 55
IGF-1 levels
LI-Igf1−/−j 88 F + 16 F 812 ± 33 F 700 ± 21 C57BL/6J ⇓ ⇓ nd nd
M+6 M 714 ± 24 M 672 ± 21

(continued)

717
Growth Hormone Axis
718
Growth Hormone Axis

Table 3 (Continued)

Human diseases Lifespan Lifespan (days)


or genetic Size (% of changes Genetic Body Insulin Tumour Stress
variants Mouse models control) (%) Mutant Control background fat sensitivity incidence resistance

50% IGF-1 IGFR variants Igf1r+/−k 90 F + 33 F 756 ± 46 F 568 ± 49 129J nd ⇓ nd ⇑


receptor activity M + 16 M 679 ± 80 M 585 ± 69
Igf1r+/−l 90 F+5 F 967 ± 29 F 923 ± 21 C57BL/6J nd ⇓ = ⇑F
M-5 M 939 ± 24 M 983 ± 21
Igf1r+/−m 90 F + 11 F 896 ± 23 F 805 ± 26 C57BL/6J nd ⇓ nd ⇑
M-3 M 803 ± 20 M 831 ± 22
BIGFRKO+/−n 90 F+8 F 888 ± 27 F 821 ± 36 C57BL/6J x ⇑ ⇓ ⇓ =
(brain specific) M + 13 M 966 ± 28 M 853 ± 43 129SV/E
Alterations in IRS2 variants Klotho 100 F1 + 19
IGF-1/Insulin transgeneo F2 + 19
signalling
M1 + 20 F1 829 ± 32 F 697 ± 45 C57BL/6J × nd ⇓M Nd ⇑
M2 + 31 F2 830 ± 29 M 715 ± 44 C3H
M1 858 ± 40
M2 936 ± 47
Irs1−/−p 70 F + 17 F 891 ± 39
M + 14
M 897 ± 41 F 763 ± 21 C57BL/6J ⇓ ⇓ nd Nd
M 786 ± 21
Irs2 +/−q 100 F + 16 F 918 ± 21 F 790 ± 14 C57BL/6J nd ⇑ nd nd
M + 18 M 900 ± 34 M 761 ± 13
Irs2 +/−r 100 F&M + 4 F&M 788 ± 17 F&M 755 ± 22 C57BL/6J nd = nd nd
Irs2−/−s 90 F - 26 F 560 ± 63 F 755 ± 23 C57BL/6J nd ⇓ nd nd
M - 84 M123 ± 20 M 767 ± 40
Comprehensive Physiology

Volume 6, April 2016


NesCreIrs2+/−t 100 F&M + 18 F&M 936 F&M 791 C57BL/6J ⇑ ⇓ nd nd
NesCreIrs2−/−t 100 F&M + 14 F&M 901
(brain specific)

P66shc+/−u 100 F&M + 7 F&M 815 ± 37 F&M 761 ± 19 129SV/Ev ⇓ = nd ⇑


P66shc−/−u F&M + 28 F&M 973 ± 37
S6K1−/−v 70 F + 20 F 950 ± 38 F 789 ± 42 C57BL/6J = nd

Volume 6, April 2016


⇓ ⇑
M+5 M 841 ± 47 M 801 ± 39
Comprehensive Physiology

Modified from Junnila, 2013 and Berryman et al., 2008.


Abreviations: F, females; M, males; nd, not determined.
a, (8) g, (1) m, (21) s, (20)
b, (4) h, (5, 6) n, (11) t, (19)
c, (8) i, (9) o, (12) u, (13)
d, (17) j, (18) p, (14, 15) v, (16)
e, (2, 7) k, (10) q, (19)
f, (2, 7) l, (3) r, (14)

1. Bartke A. Is growth hormone deficiency a beneficial adaptation to aging? Evidence from experimental animals. Trends Endocrinol Metab 14: 340-344, 2003.
2. Berryman DE, List EO, and Kopchick JJ. Repression of GH signaling: one extended life to live! Aging (Albany NY) 5: 723-724, 2013.
3. Bokov AF, Garg N, Ikeno Y, Thakur S, Musi N, DeFronzo RA, Zhang N, Erickson RC, Gelfond J, Hubbard GB, Adamo ML, and Richardson A. Does reduced IGF-1R signaling in Igf1r+/−
mice alter aging? PLoS One 6: e26891, 2011.
4. Brown-Borg HM, Borg KE, Meliska CJ, and Bartke A. Dwarf mice and the ageing process. Nature 384: 33, 1996.
5. Conover CA. Role of PAPP-A in aging and age-related disease. Exp Gerontol 48: 612-613, 2013.
6. Conover CA, and Bale LK. Loss of pregnancy-associated plasma protein A extends lifespan in mice. Aging Cell 6: 727-729, 2007.
7. Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, and Kopchick JJ. Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights,
insulin, and insulin-like growth factor I levels and increased life span. Endocrinology 144: 3799-3810, 2003.
8. Flurkey K, Papaconstantinou J, Miller RA, and Harrison DE. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl
Acad Sci U S A 98: 6736-6741, 2001.
9. Gong Z, Kennedy O, Sun H, Wu Y, Williams GA, Klein L, Cardoso L, Matheny RW, Jr., Hubbard GB, Ikeno Y, Farrar RP, Schaffler MB, Adamo ML, Muzumdar RH, and Yakar S. Reductions
in serum IGF-1 during aging impair health span. Aging Cell 13: 408-418, 2014.
10. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, and Le Bouc Y. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421:
182-187, 2003.
11. Kappeler L, De Magalhaes Filho C, Dupont J, Leneuve P, Cervera P, Perin L, Loudes C, Blaise A, Klein R, Epelbaum J, Le Bouc Y, and Holzenberger M. Brain IGF-1 receptors control mammalian
growth and lifespan through a neuroendocrine mechanism. PLoS Biol 6: e254, 2008.
12. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP,
and Kuro-o M. Suppression of aging in mice by the hormone Klotho. Science 309: 1829-1833, 2005.
13. Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP, Lanfrancone L, and Pelicci PG. The p66shc adaptor protein controls oxidative stress response and life span in mammals.
Nature 402: 309-313, 1999.
14. Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M, Clements M, Ramadani F, Okkenhaug K, Schuster E, Blanc E, Piper MD, Al-Qassab H, Speakman JR, Carmignac D, Robinson
IC, Thornton JM, Gems D, Partridge L, and Withers DJ. Evidence for lifespan extension and delayed age-related biomarkers in insulin receptor substrate 1 null mice. FASEB J 22: 807-818, 2008.
15. Selman C, Partridge L, and Withers DJ. Replication of extended lifespan phenotype in mice with deletion of insulin receptor substrate 1. PLoS One 6: e16144, 2011.
16. Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury AI, Claret M, Al-Qassab H, Carmignac D, Ramadani F, Woods A, Robinson IC, Schuster E, Batterham RL, Kozma SC, Thomas
G, Carling D, Okkenhaug K, Thornton JM, Partridge L, Gems D, and Withers DJ. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science 326: 140-144, 2009.
17. Sun LY, Spong A, Swindell WR, Fang Y, Hill C, Huber JA, Boehm JD, Westbrook R, Salvatori R, and Bartke A. Growth hormone-releasing hormone disruption extends lifespan and regulates
response to caloric restriction in mice. Elife 2: e01098, 2013.
18. Svensson J, Sjogren K, Faldt J, Andersson N, Isaksson O, Jansson JO, and Ohlsson C. Liver-derived IGF-I regulates mean life span in mice. PLoS One 6: e22640, 2011.
19. Taguchi A, Wartschow LM, and White MF. Brain IRS2 signaling coordinates life span and nutrient homeostasis. Science 317: 369-372, 2007.
20. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, Bonner-Weir S, and White MF. Disruption of IRS-2 causes type 2 diabetes in mice.
Nature 391: 900-904, 1998.
21. Xu J, Gontier G, Chaker Z, Lacube P, Dupont J, and Holzenberger M. Longevity effect of IGF-1R(+/−) mutation depends on genetic background-specific receptor activation. Aging Cell 13:
19-28, 2014.

719
Growth Hormone Axis
Growth Hormone Axis Comprehensive Physiology

Two dwarf mouse lines with decreased GH action exhibit short stature and elevated IGF-1, suggesting IGF-1 resistance
strikingly different phenotypes in term of life span extension: (492, 522).
the GH receptor-invalidated (Ghr−/−) mouse is completely Post IGF-1 and insulin receptor signaling has also been
resistant to GH action and long-lived while another dwarf targeted on insulin receptor substrates (IRS), which are dock-
mouse line expressing a GH-R antagonist (GHA) transgene ing proteins between the membrane receptors and their mul-
is only partially resistant to GH. Compared to the Ghr−/−, life tiple intracellular pathways. Out of the 4 IRS, IRS1, and
span extension in this mouse is not as significant. Circulating IRS2 are the major insulin receptor substrates leading to glu-
IGF-1 levels are less reduced in the GHA when compared cose homeostasis, and have distinct and overlapping roles in
to the GH-R-/- mouse, and if glucose homeostasis is mod- diverse organs. The majority of the published literature in
estly improved in young GHA animals, it deteriorates with this field suggests that IRS1 is the major substrate leading to
age. In humans, GH resistance corresponds to the Laron syn- stimulation of glucose transport in muscle and adipose tis-
drome (182,290,291,443), caused by homozygous mutations sues, whereas in liver, IRS1 and IRS2 have complementary
in GHR or GHR-signaling molecules. Laron adult heights roles in insulin signaling and metabolism. Constitutive dele-
vary from 106 to 142 cm and they are obese. Protection from tion of IRS1 extends lifespan in IRS-1 knockout (Irs1−/−)
diabetes is observed in a large Ecuatorian cohort but not in mice (467, 468) while IRS-2 knockout (Irs2−/−) mice are
the original Mediterranean one. No change in lifespan in this diabetic and considerably short lived (467). Lifespan is not
population has been reported as yet. affected in Irs1 heterozygous (Irs1+/−) mice (467) while Irs2
Growth hormone excess in GH overexpressing transgenic heterozygous (Irs2+/−) mice (which have normal to improved
mice (29,398,421), as is seen in acromegalic patients bearing insulin sensitivity (500,580) display extended (500) or normal
GH hypersecreting adenomas (85), induces gigantism, lean- (467) longevity. In Humans, one study of an Italian popula-
ness, insulin resistance and shortens life span. Decreasing tion revealed that individuals heterozygous or homozygous
GH and normalizing IGF-1 levels, by surgery, radiotherapy for a common IRS2 gene variant, Gly1057Asp, present in
or pharmacological treatment with somatostatin analogues about a third of the population, have a higher probability of
or the GH antagonist pegvisomant, reduces the mortality reaching old age (96-104 years) than bearers of the wild-
rate of acromegalic patients to that of the normal population type allele (27). When tested together with two other com-
(37, 124, 209). mon alleles, G/A-IGF1R and the mitochondrial uncoupling
In contrast to mice lacking GHRH, GH or the GH protein 2 Ala/Val-UCP2, the AAV allele combination was
receptor which are viable, neonatal deaths often occur in associated with a decrease in all-cause mortality risk and a
IGF-1 knockout (Igf1−/−) dwarf (60% of normal size) higher probability to reach extreme old age as well as lower
mice. Neonatal death ratio depends on the mouse genetic homeostatic model assessment ratio (HOMA-R), a measure
background (90% in 129SVX 129SV to 32% in MF1x129SV of insulin resistance, higher respiratory quotient, and resting
crosses). Null mutants for the Igf1r gene invariably die at metabolic rate (27).
birth as a consequence of respiratory failure, with a more Taken altogether, this complex set of data raises the ques-
severe growth deficiency (45% of normal birth weight) tion of the tissue targets for IGF-1 in extending life span. In
(307). Therefore, several strategies were designed to assess this respect, cardiac-specific overexpression of IGF-1 posi-
the effects of IGF-1 deficiency in nonlethal mouse mutants. tively affected mouse median but not maximal lifespan (298).
Pregnancy-associated plasma protein A (PAPP-A) is a Conversely, neuron-specific reduction of the IGF-1/insulin
metalloproteinase that degrade inhibitory IGF-Binding receptor ortholog Daf-2 in C. elegans (581) and brain-specific
Proteins (IGFBP), particularly IGFBP4, in the pericellular reduction of IGF1-R1 (248) or IRS2 (500) in mice were
environment; thereby increasing IGF-1 bioavailability reported to increase maximal life span (again with a much
without changing IGF-I levels. Genetic deletion of PAPP-A bigger effect in worms than mammals). Moreover, the invali-
extends mean and maximum lifespans by 30% to 40%, with dation of Surfeit locus protein 1 (SURF1), a 30 kDa hydropho-
no reduction in food intake or secondary endocrine abnor- bic protein embedded in the inner membrane of mitochon-
malities and reduced incidence of spontaneous tumors (101). dria, also results in markedly prolonged median lifespan, and
In contrast, when 75% decreases in circulating IGF-1 levels complete protection from calcium-dependent neurotoxicity
were achieved by liver-specific invalidation of IGF-1 in two induced by kainic acid (125). However, the brain is not the
mouse strains (LID−/− (180) and LI-Igf1−/− (497) mice), only target organ involved since extended longevity is also
modest (<20%), sex-dependent and contradictory modifica- observed in mice lacking the insulin receptor in adipose tissue
tions in life spans were reported. Contradictory reports also (50). The kidney may also be involved since Klotho protein,
exist for constitutive IGF-1r heterozygous (Igf1r+/−) mice, a transmembrane protein with homology to B-glycosidases,
which display a 50% or no reduction in circulating IGF-1 is principally expressed in the distal tubules (and the choroid
levels (52,217,585). The effects on life span appear to depend plexus). Klotho may also acts as a hormone and may bind and
on the conditions in which these mice are kept and crossed. repress IGF-1 and insulin intracellular signals (281). Klotho
However, human IGF1R variants with a higher frequency in overexpression in mice extends lifespan but in contrast to
centenarians (2.3%) than controls (0.3%) have been reported previously discussed mouse models with increased lifespan,
to suppress IGF1 signaling and female carriers have slightly Klotho transgenic mice are insulin resistant (281).

720 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

In summary, corresponding or similar mutations in the number of neuropeptides, neurohormones, and peripheral fac-
GHRH/GH/IGF-1 pathway have been identified in mouse tors including gut hormones and energy substrates. While
and Man, but whether these mutations alter longevity has impacting the amount of GH released, these factors also
yet to be confirmed in the latter species. At any rate, several determine the patterning of GH release, and this change in
of these mouse and human mutations with reduced activity patterning may affect the actions of GH more so than changes
of the GH/IGF-1 axis appear protective against cancer and in the amount of GH released. To this extent, future studies
diabetes mellitus, two major ageing-related morbidities. The must consider measures of irregularity and pulse frequency,
activity of the GH-IGF-1 axis is also decreased by calorie particularly in animal models for which these data are scarce.
restriction (CR), a powerful intervention that slows the age- In many regards, neuronal regulators that couple GH release
ing process and decreases mortality in a range of organisms, with food intake (including NPY) are emerging as suitable
including rhesus monkeys (98). Deletion of ribosomal S6 pro- candidates. Given the developing role of GH as a critical
tein kinase 1 (S6K1), a component of the nutrient-responsive regulator of long-term energy homoeostasis, and emerging
mTOR (mammalian target of rapamycin) signaling pathway, evidence of neuronal regulators of food intake in the con-
downstream of IRS1 signaling, lead to increased life span and trol of GH release, studies that couple GH release with food
resistance to age-related pathologies, such as bone, immune, intake may further define mechanisms of GH action that pre-
and motor dysfunction and loss of insulin sensitivity (469). dict the use of energy during metabolic extremes. We must
Life extension is also present in p66shc−/− mice (342). The however acknowledge that the context under which GH mod-
p66shc isoform of SHC-transforming protein 1 has been sug- ulates energy homeostasis in the various animal models often
gested to couple IRS1 and the S6 kinase. used to define GH regulation and action differs according to
species; therefore, impacting our capacity to directly translate
observations into human physiology. There is much to gain
Conclusion through recognizing these differences, and through the devel-
opment of a greater physiological understanding behind these
Assessment of the complex interactions that define GH release differences.
has seen the evolution of models that predict and interpret As listed throughout this article, the release of GH changes
GH output under particular physiological or pathophysiolog- considerably relative to metabolic demand. This is clearly
ical conditions. Considerable insights now help to define the illustrated in female physiology, wherein altered metabolic
actions of GH in growth, metabolism, and longevity, and the needs throughout the ovulatory cycle, in pregnancy and in
role of the neuroendocrine system in sustaining these fun- lactation may predict GH output (as detailed earlier). While
damental processes of life. These led to significant clinical dramatic changes in the hormonal milieu specific to these dif-
improvement for the therapy of acromegaly through long ferent reproductive states contribute to increased (i.e., estra-
acting somatostatin analogs (379) and GH receptor antag- diol) or decreased (i.e., placental-derived GH) GH output, the
onism (277). More recently, a botulinum neurotoxin-derived physiological context of increased or decreased GH release
targeted secretion inhibitor has been designed to target pitu- remains mostly unknown. Should we hope to understand this,
itary somatotroph cells and suppress GH secretion (476). This current measures must be extended to define the mechanisms
recombinant protein contains a modified GHRH domain and of actions of GH under these and other metabolic extremes,
the endopeptidase domain of botulinum toxin serotype D, thus and through studying the interactions of GH with other crit-
targeting the GHRH receptor and depleting vesicle-associated ical regulators of energy homoeostasis (including insulin).
membrane protein 2 (VAMP2) a SNARE protein involved in Indeed, an extension in lifespan following genetic alterations
GH exocytosis. Beyond the GH axis, somatostatin receptor that compromise GH signaling is often associated with an
imaging is also of interest for diagnosis and therapy of neu- improved capacity to tolerate life-long pressures on energy
roendocrine tumors (542). homeostasis. Only by understanding the mechanisms that
While significant progress was made in defining the neu- predict GH release throughout life, and relative to other crit-
ronal processes that govern GH output, a complete overview ical regulators of energy homeostasis, will we define strate-
of these interactions is still lacking. In this pursuit we gies for improving health-span and longevity. Indeed, our
must direct our attention to the neurovascular unit of the emerging understanding of the role of GH in regulating long-
hypothalamo-pituitary complex, as has been done for the reg- term energy homeostasis, and by proxy health-span, offers the
ulation of the gonadotropic axis. Development of a greater greatest incentive yet in defining mechanisms of GH release.
understanding of the control and release of somatostatin and
GHRH will reveal the cause for the seemingly discordant
release of peripheral GH output relative to somatostatin and References
GHRH content in pituitary portal vasculature. By making this
1. Abe H, Molitch ME, Van Wyk JJ, Underwood LE. Human growth hor-
a research priority, we may perhaps extend our list of primary mone and somatomedin C suppress the spontaneous release of growth
regulators of GH release beyond somatostatin and GHRH. hormone in unanesthetized rats. Endocrinology 113: 1319-1324, 1983.
2. Accorsi PA, Munno A, Gamberoni M, Viggiani R, De Ambrogi M,
As is detailed throughout this article, mechanisms that Tamanini C, Seren E. Role of leptin on growth hormone and prolactin
control GH release are varied, and involve a very large secretion by bovine pituitary explants. J Dairy Sci 90: 1683-1691, 2007.

Volume 6, April 2016 721


Growth Hormone Axis Comprehensive Physiology

3. Aguiar-Oliveira MH, Oliveira FT, Pereira RM, Oliveira CR, Blackford 25. Barber MC, Clegg RA, Finley E, Vernon RG, Flint DJ. The role of
A, Valenca EH, Santos EG, Gois-Junior MB, Meneguz-Moreno RA, growth hormone, prolactin and insulin-like growth factors in the reg-
Araujo VP, Oliveira-Neto LA, Almeida RP, Santos MA, Farias NT, Sil- ulation of rat mammary gland and adipose tissue metabolism during
veira DC, Cabral GW, Calazans FR, Seabra JD, Lopes TF, Rodrigues lactation. J Endocrinol 135: 195-202, 1992.
EO, Porto LA, Oliveira IP, Melo EV, Martari M, Salvatori R. Longevity 26. Barber MC, Travers MT, Finley E, Flint DJ, Vernon RG. Growth-
in untreated congenital growth hormone deficiency due to a homozy- hormone-prolactin interactions in the regulation of mammary and
gous mutation in the GHRH receptor gene. J Clin Endocrinol Metab adipose-tissue acetyl-CoA carboxylase activity and gene expression
95: 714-721, 2010. in lactating rats. Biochem J 285(Pt 2): 469-475, 1992.
4. Aguila MC, Marubayashi U, McCann SM. The effect of galanin on 27. Barbieri M, Boccardi V, Esposito A, Papa M, Vestini F, Rizzo MR,
growth hormone-releasing factor and somatostatin release from median Paolisso G. A/ASP/VAL allele combination of IGF1R, IRS2, and UCP2
eminence fragments in vitro. Neuroendocrinology 56: 889-894, 1992. genes is associated with better metabolic profile, preserved energy
5. Aguila MC, McCann SM. Growth hormone increases somatostatin expenditure parameters, and low mortality rate in longevity. Age 34:
release and messenger ribonucleic acid levels in the rat hypothalamus. 235-245, 2012.
Brain Res 623: 89-94, 1993. 28. Bartke A. Can growth hormone (GH) accelerate aging? Evidence from
6. Al-Massadi O, Gabellieri E, Trujillo ML, Senaris R, Pagotto U, Pasquali GH-transgenic mice. Neuroendocrinology 78: 210-216, 2003.
R, Casanueva FF, Seoane LM. Peripheral endocannabinoid system- 29. Bartke A. Is growth hormone deficiency a beneficial adaptation to
mediated actions of rimonabant on growth hormone secretion are aging? Evidence from experimental animals. Trends Endocrinol Metab
ghrelin-dependent. J Neuroendocrinol 22: 1127-1136, 2010. 14: 340-344, 2003.
7. Al-Massadi O, Trujillo ML, Senaris R, Pardo M, Castelao C, Casanueva 30. Bartke A, Coschigano K, Kopchick J, Chandrashekar V, Mattison J,
FF, Seoane LM. The vagus nerve as a regulator of growth hormone Kinney B, Hauck S. Genes that prolong life: Relationships of growth
secretion. Regul Pept 166: 3-8, 2011. hormone and growth to aging and life span. J Gerontol A Biol Sci Med
8. Alford FP, Baker HW, Burger HG, De Kretser DM, Hudson B, Johns Sci 56: B340-349, 2001.
MW, Masterton JP, Patel YC, Rennie GC. Temporal patterns of inte- 31. Bassett NS, Gluckman PD. Pulsatile growth hormone secretion in the
grated plasma hormone levels during sleep and wakefulness. I. Thyroid- ovine fetus and neonatal lamb. J Endocrinol 109: 307-312, 1986.
stimulating hormone, growth hormone and cortisol. J Clin Endocrinol 32. Bauman DE, Vernon RG. Effects of exogenous bovine somatotropin
Metab 37: 841-847, 1973. on lactation. Annu Rev Nutr 13: 437-461, 1993.
9. Alvarez CV, Mallo F, Burguera B, Cacicedo L, Dieguez C, Casanueva 33. Bedard K, Strecko J, Theriault K, Bedard J, Veyrat-Durebex C, Gau-
FF. Evidence for a direct pituitary inhibition by free fatty-acids in vivo dreau P. Effects of a high-glucose environment on the pituitary growth
growth-hormone responses to growth hormone-releasing hormone in hormone-releasing hormone receptor: Type 1 diabetes compared with
the rat. Neuroendocrinology 53: 185-189, 1991. in vitro glucotoxicity. Am J Physiol Endocrinol Metab 294: E740-751,
10. Amiel SA, Sherwin RS, Hintz RL, Gertner JM, Press CM, Tamborlane 2008.
WV. Effect of diabetes and its control on insulin-like growth factors in 34. Belfort MB, Brown RS. Fetal and postnatal growth: Mechanisms, con-
the young subject with type I diabetes. Diabetes 33: 1175-1179, 1984. sequences and controversies. Curr Opin Endocrinol Diabetes Obes 14:
11. Anderson SM, Wideman L, Patrie JT, Weltman A, Bowers CY, Veld- 1-2, 2007.
huis JD. E2 supplementation selectively relieves GH’s autonegative 35. Bellantoni MF, Harman SM, Cho DE, Blackman MR. Effects of
feedback on GH-releasing peptide-2-stimulated GH secretion. J Clin progestin-opposed transdermal estrogen administration on growth hor-
Endocrinol Metab 86: 5904-5911, 2001. mone and insulin-like growth factor-I in postmenopausal women of
12. Andreotti AC, Lanzi R, Manzoni MF, Caumo A, Moreschi A, Pontiroli different ages. J Clin Endocrinol Metab 72: 172-178, 1991.
AE. Acute pharmacological blockade of lipolysis normalizes nocturnal 36. Bellantoni MF, Vittone J, Campfield AT, Bass KM, Harman SM, Black-
growth hormone levels and pulsatility in obese subjects. Metabolism man MR. Effects of oral versus transdermal estrogen on the growth
43: 1207-1213, 1994. hormone/insulin-like growth factor I axis in younger and older post-
13. Antoni FA, Kanyicska B, Mezey E, Makara GB. Neonatal treatment menopausal women: A clinical research center study. J Clin Endocrinol
with monosodium-L-glutamate: Differential effects on growth hormone Metab 81: 2848-2853, 1996.
and prolactin release induced by morphine. Neuroendocrinology 35: 37. Ben-Shlomo A, Sheppard MC, Stephens JM, Pulgar S, Melmed S.
231-235, 1982. Clinical, quality of life, and economic value of acromegaly disease
14. Arai R, Calas A. Ultrastructural localization of galanin immunoreac- control. Pituitary 14: 284-294, 2011.
tivity in the rat median eminence. Brain Res 562: 339-343, 1991. 38. Bennett PA, Levy A, Sophokleous S, Robinson IC, Lightman SL.
15. Ariyasu H, Iwakura H, Yamada G, Kanamoto N, Bando M, Kohno K, Hypothalamic GH receptor gene expression in the rat: Effects of altered
Sato T, Kojima M, Nakao K, Kangawa K, Akamizu T. A postwean- GH status. J Endocrinol 147: 225-234, 1995.
ing reduction in circulating ghrelin temporarily alters growth hormone 39. Berelowitz M, Firestone SL, Frohman LA. Effects of growth hormone
(GH) responsiveness to GH-releasing hormone in male mice but does excess and deficiency on hypothalamic somatostatin content and release
not affect somatic growth. Endocrinology 151: 1743-1750, 2010. and on tissue somatostatin distribution. Endocrinology 109: 714-719,
16. Arvat E, Giordano R, Gianotti L, Broglio F, Camanni F, Ghigo E. 1981.
Neuroendocrinology of the human growth hormone-insulin-like growth 40. Berelowitz M, Szabo M, Frohman LA, Firestone S, Chu L, Hintz RL.
factor I axis during ageing. Growth Horm IGF Res 9(Suppl A): 111-115, Somatomedin-C mediates growth hormone negative feedback by effects
1999. on both the hypothalamus and the pituitary. Science 212: 1279-1281,
17. Asakawa A, Inui A, Fujimiya M, Sakamaki R, Shinfuku N, Ueta Y, 1981.
Meguid MM, Kasuga M. Stomach regulates energy balance via acylated 41. Bermann M, Jaffe CA, Tsai W, DeMott-Friberg R, Barkan AL. Negative
ghrelin and desacyl ghrelin. Gut 54: 18-24, 2005. feedback regulation of pulsatile growth hormone secretion by insulin-
18. Asakawa A, Inui A, Kaga T, Katsuura G, Fujimiya M, Fujino MA, like growth factor I. Involvement of hypothalamic somatostatin. J Clin
Kasuga M. Antagonism of ghrelin receptor reduces food intake and Invest 94: 138-145, 1994.
body weight gain in mice. Gut 52: 947-952, 2003. 42. Bertherat J, Berod A, Normand E, Bloch B, Rostenef W, Kordon C,
19. Asplin CM, Faria AC, Carlsen EC, Vaccaro VA, Barr RE, Iranmanesh Epelbaum J. Somatostatin depletion by cysteamine increases somato-
A, Lee MM, Veldhuis JD, Evans WS. Alterations in the pulsatile mode statin binding and growth hormone-releasing factor messenger ribonu-
of growth hormone release in men and women with insulin-dependent cleic Acid in the arcuate nucleus. J Neuroendocrinol 3: 115-118,
diabetes mellitus. J Clin Endocrinol Metab 69: 239-245, 1989. 1991.
20. Astrom C. Interaction between sleep and growth hormone. Evaluated 43. Bertherat J, Timsit J, Bluet-Pajot MT, Mercadier JJ, Gourdji D, Kordon
by manual polysomnography and automatic power spectrum analysis. C, Epelbaum J. Chronic growth hormone (GH) hypersecretion induces
Acta Neurol Scand 92: 281-296, 1995. reciprocal and reversible changes in mRNA levels from hypothalamic
21. Astrom C, Pedersen SA, Lindholm J. The influence of growth hormone GH-releasing hormone and somatostatin neurons in the rat. J Clin Invest
on sleep in adults with growth hormone deficiency. Clin Endocrinol 91: 1783-1791, 1993.
(Oxf) 33: 495-500, 1990. 44. Besset A, Bonardet A, Billiard M, Descomps B, de Paulet AC, Pas-
22. Baker HW, Best JB, Burger HG, Cameron DP. Plasma human growth souant P. Circadian patterns of growth hormone and cortisol secretions
hormone levels in response to meals: A reappraisal. Aust J Exp Biol in narcoleptic patients. Chronobiologia 6: 19-31, 1979.
Med Sci 50: 715-724, 1972. 45. Besson A, Salemi S, Gallati S, Jenal A, Horn R, Mullis PS, Mullis PE.
23. Balthasar N, Mery P-F, Magoulas CB, Mathers KE, Martin A, Mollard Reduced longevity in untreated patients with isolated growth hormone
P, Robinson ICAF. Growth hormone-releasing hormone (GHRH) neu- deficiency. J Clin Endocrinol Metab 88: 3664-3667, 2003.
rons in GHRH-enhanced green fluorescent protein transgenic mice: A 46. Bivens CH, Lebovitz HE, Feldman JM. Inhibition of hypoglycemia-
ventral hypothalamic network. Endocrinology 144: 2728-2740, 2003. induced growth hormone secretion by the serotonin antagonists cypro-
24. Barbarino A, Corsello SM, Della Casa S, Tofani A, Sciuto R, Rota CA, heptadine and methysergide. N Engl J Med 289: 236-239, 1973.
Bollanti L, Barini A. Corticotropin-releasing hormone inhibition of 47. Bluet-Pajot MT, Bertherat J, Epelbaum J, Kordon C. Neural and pitu-
growth hormone-releasing hormone-induced growth hormone release itary mechanisms involved in growth hormone regulation. J Pediatr
in man. J Clin Endocrinol Metab 71: 1368-1374, 1990. Endocrinol 6: 357-369, 1993.

722 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

48. Bluet-Pajot MT, Durand D, Kordon C. Influence of streptozotocin- intracellular signaling systems. Role of adenosine. Neuroendocrinology
induced diabetes on growth hormone secretion in the rat. Neuroen- 79: 13-25, 2004.
docrinology 36: 307-309, 1983. 73. Casanueva FF, Villanueva L, Cabranes JA, Cabezas-Cerrato J,
49. Bluet-Pajot MT, Mounier F, Durand D, Kordon C. Involvement of Fernandez-Cruz A. Cholinergic mediation of growth hormone secre-
dopamine D1 receptors in the control of growth hormone secretion in tion elicited by arginine, clonidine, and physical exercise in man. J Clin
the rat. J Endocrinol 127: 191-196, 1990. Endocrinol Metab 59: 526-530, 1984.
50. Bluher M, Kahn BB, Kahn CR. Extended longevity in mice lacking the 74. Casanueva FF, Villanueva L, Dieguez C, Diaz Y, Cabranes JA, Szoke
insulin receptor in adipose tissue. Science 299: 572-574, 2003. B, Scanlon MF, Schally AV, Fernandezcruz A. Free fatty-acids block
51. Boden G, Chen X, Mozzoli M, Ryan I. Effect of fasting on serum leptin growth-hormone (GH) releasing hormone-stimulated GH secretion in
in normal human subjects. J Clin Endocrinol Metab 81: 3419-3423, man directly at the pituitary. J Clin Endocrinol Metab 65: 634-642,
1996. 1987.
52. Bokov AF, Garg N, Ikeno Y, Thakur S, Musi N, DeFronzo RA, Zhang 75. Cataldi M, Magnan E, Guillaume V, Dutour A, Conte-Devolx B, Lom-
N, Erickson RC, Gelfond J, Hubbard GB, Adamo ML, Richardson A. bardi G, Oliver C. Relationship between hypophyseal portal GHRH
Does reduced IGF-1R signaling in Igf1r+/− mice alter aging? PLoS and somatostatin and peripheral GH levels in the conscious sheep.
One 6: e26891, 2011. J Endocrinol Invest 17: 717-722, 1994.
53. Bonnefont X, Lacampagne A, Sanchez-Hormigo A, Fino E, Creff A, 76. Catzeflis C, Pierroz DD, Rohner-Jeanrenaud F, Rivier JE, Sizonenko
Mathieu MN, Smallwood S, Carmignac D, Fontanaud P, Travo P, PC, Aubert ML. Neuropeptide Y administered chronically into the lat-
Alonso G, Courtois-Coutry N, Pincus SM, Robinson IC, Mollard P. eral ventricle profoundly inhibits both the gonadotropic and the soma-
Revealing the large-scale network organization of growth hormone- totropic axis in intact adult female rats. Endocrinology 132: 224-234,
secreting cells. Proc Natl Acad Sci U S A 102: 16880-16885, 2005. 1993.
54. Bowers CY. GH releasing peptides–structure and kinetics. J Pediatr 77. Cella SG, Arce VM, Pieretti F, Locatelli V, Settembrini BP, Muller
Endocrinol 6: 21-31, 1993. EE. Combined administration of growth-hormone-releasing hormone
55. Bowers CY. Growth hormone releasing peptides. In: Kostyo JL, editor. and clonidine restores defective growth hormone secretion in old dogs.
Handbook of Physiology. New York, Oxford: Oxford University Press Neuroendocrinology 57: 432-438, 1993.
for the American Physiology Society, 1999, pp. 267-297. 78. Cella SG, Luceri M, Cattaneo L, Torsello A, Muller EE. Somatostatin
56. Boyd AE, 3rd, Lebovitz HE, Pfeiffer JB. Stimulation of human-growth- withdrawal as generator of pulsatile GH release in the dog: A possible
hormone secretion by L-dopa. N Engl J Med 283: 1425-1429, 1970. tool to evaluate the endogenous GHRH tone? Neuroendocrinology 63:
57. Branchek TA, Smith KE, Gerald C, Walker MW. Galanin receptor 481-488, 1996.
subtypes. Trends Pharmacol Sci 21: 109-117, 2000. 79. Cella SG, Moiraghi V, Minuto F, Barreca A, Cocchi D, De Gen-
58. Brandenberger G, Gronfier C, Chapotot F, Simon C, Piquard F. Effect naro Colona V, Reina G, Muller EE. Prolonged fasting or clonidine
of sleep deprivation on overall 24 h growth-hormone secretion. Lancet can restore the defective growth hormone secretion in old dogs. Acta
356: 1408, 2000. endocrinologica 121: 177-184, 1989.
59. Bray MJ, Vick TM, Shah N, Anderson SM, Rice LW, Iranmanesh 80. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role
A, Evans WS, Veldhuis JD. Short-term estradiol replacement in post- of falling leptin levels in the neuroendocrine and metabolic adaptation
menopausal women selectively mutes somatostatin’s dose-dependent to short-term starvation in healthy men. J Clin Invest 111: 1409-1421,
inhibition of fasting growth hormone secretion. J Clin Endocrinol 2003.
Metab 86: 3143-3149, 2001. 81. Chan JL, Williams CJ, Raciti P, Blakeman J, Kelesidis T, Kelesidis
60. Brazeau P, Vale W, Burgus R, Ling N, Butcher M, Rivier J, Guillemin R. I, Johnson ML, Thorner MO, Mantzoros CS. Leptin does not mediate
Hypothalamic polypeptide that inhibits the secretion of immunoreactive short-term fasting-induced changes in growth hormone pulsatility but
pituitary growth hormone. Science 179: 77-79, 1973. increases IGF-I in leptin deficiency states. J Clin Endocrinol Metab 93:
61. Briard N, Rico-Gomez M, Guillaume V, Sauze N, Vuaroqueaux V, 2819-2827, 2008.
Dadoun F, Le Bouc Y, Oliver C, Dutour A. Hypothalamic medi- 82. Chan YY, Clifton DK, Steiner RA. Role of NPY neurones in GH-
ated action of free fatty acid on growth hormone secretion in sheep. dependent feedback signalling to the brain. Horm Res 45 (Suppl 1):
Endocrinology 139: 4811-4819, 1998. 12-14, 1996.
62. Broberger C, Landry M, Wong H, Walsh JN, Hokfelt T. Subtypes Y1 83. Chan YY, Grafstein-Dunn E, Delemarre-van de Waal HA, Burton KA,
and Y2 of the neuropeptide Y receptor are respectively expressed in Clifton DK, Steiner RA. The role of galanin and its receptor in the
pro-opiomelanocortin- and neuropeptide-Y-containing neurons of the feedback regulation of growth hormone secretion. Endocrinology 137:
rat hypothalamic arcuate nucleus. Neuroendocrinology 66: 393-408, 5303-5310, 1996.
1997. 84. Chan YY, Steiner RA, Clifton DK. Regulation of hypothalamic
63. Broglio F, Prodam F, Riganti F, Gottero C, Destefanis S, Granata R, neuropeptide-Y neurons by growth hormone in the rat. Endocrinol-
Muccioli G, Abribat T, van der Lely AJ, Ghigo E. The continuous ogy 137: 1319-1325, 1996.
infusion of acylated ghrelin enhances growth hormone secretion and 85. Chanson P, Salenave S. Acromegaly. Orphanet J Rare Dis 3: 17,
worsens glucose metabolism in humans. J Endocrinol Invest 31: 788- 2008.
794, 2008. 86. Chartrel N, Alvear-Perez R, Leprince J, Iturrioz X, Reaux-Le Goazigo
64. Brown WA, Krieger DT, Van Woert MH, Ambani LM. Dissociation A, Audinot V, Chomarat P, Coge F, Nosjean O, Rodriguez M, Galizzi
of growth hormone and cortisol release following apomorphine. J Clin JP, Boutin JA, Vaudry H, Llorens-Cortes C. Comment on “Obestatin, a
Endocrinol Metab 38: 1127-1130, 1974. peptide encoded by the ghrelin gene, opposes ghrelin’s effects on food
65. Bruni JF, Meites J. Effects of cholinergic drugs on growth hormone intake”. Science 315: 766; author reply 766, 2007.
release. Life Sci 23: 1351-1357, 1978. 87. Chen G, van den Pol AN. Multiple NPY receptors coexist in pre-
66. Bruni JF, Van Vugt D, Marshall S, Meites J. Effects of naloxone, and postsynaptic sites: Inhibition of GABA release in isolated self-
morphine and methionine enkephalin on serum prolactin, luteinizing innervating SCN neurons. J Neurosci 16: 7711-7724, 1996.
hormone, follicle stimulating hormone, thyroid stimulating hormone 88. Cheng K, Chan WW, Butler B, Wei L, Schoen WR, Wyvratt MJ, Jr.,
and growth hormone. Life Sci 21: 461-466, 1977. Fisher MH, Smith RG. Stimulation of growth hormone release from
67. Burgevin MC, Loquet I, Quarteronet D, Habert-Ortoli E. Cloning, phar- rat primary pituitary cells by L-692,429, a novel non-peptidyl GH
macological characterization, and anatomical distribution of a rat cDNA secretagogue. Endocrinology 132: 2729-2731, 1993.
encoding for a galanin receptor. J Mol Neurosci 6: 33-41, 1995. 89. Cheng K, Chan WW, Butler B, Wei L, Smith RG. A novel non-peptidyl
68. Burton KA, Kabigting EB, Clifton DK, Steiner RA. Growth hormone growth hormone secretagogue. Horm Res 40: 109-115, 1993.
receptor messenger ribonucleic acid distribution in the adult male rat 90. Childs GV, Akhter N, Haney A, Syed M, Odle A, Cozart M, Brodrick Z,
brain and its colocalization in hypothalamic somatostatin neurons. Gaddy D, Suva LJ, Akel N, Crane C, Benes H, Charlesworth A, Luque
Endocrinology 131: 958-963, 1992. R, Chua S, Kineman RD. The somatotrope as a metabolic sensor:
69. Burton KA, Kabigting EB, Steiner RA, Clifton DK. Identification of Deletion of leptin receptors causes obesity. Endocrinology 152: 69-81.
target cells for growth hormone’s action in the arcuate nucleus. Am J 91. Chomczynski P, Downs TR, Frohman LA. Feedback regulation of
Physiol 269: E716-722, 1995. growth hormone (GH)-releasing hormone gene expression by GH in
70. Burton KA, Steiner RA, Clifton DK. Double-label in situ hybridization rat hypothalamus. Mol Endocrinol 2: 236-241, 1988.
confirms that the GRF gene and the GH receptor gene are coexpressed 92. Chowen JA, Frago LM, Argente J. The regulation of GH secretion by
in a subset of neurons in the arcuate nucleus. In: 74th Annual Meeting sex steroids. Eur J Endocrinol 151(Suppl 3): U95-100, 2004.
Of the Endocrine Society. San Antonio: The Endocrine Society Press, 93. Clark RG, Carlsson LM, Trojnar J, Robinson IC. The effects of a
1992. growth hormone-releasing Peptide and growth hormone-releasing fac-
71. Cahill GF, Jr., Herrera MG, Morgan AP, Soeldner JS, Steinke J, Levy tor in conscious and anaesthetized rats. J Neuroendocrinol 1: 249-255,
PL, Reichard GA, Jr., Kipnis DM. Hormone-fuel interrelationships 1989.
during fasting. J Clin Invest 45: 1751-1769, 1966. 94. Clark RW, Schmidt HS, Malarkey WB. Disordered growth hormone
72. Carreira MC, Camiña JP, Smith RG, Casanueva FF. Agonist-specific and prolactin secretion in primary disorders of sleep. Neurology 29:
coupling of growth hormone secretagogue receptor type 1a to different 855-861, 1979.

Volume 6, April 2016 723


Growth Hormone Axis Comprehensive Physiology

95. Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto 119. De Marinis L, Bianchi A, Mancini A, Gentilella R, Perrelli M, Giampi-
D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, etro A, Porcelli T, Tilaro L, Fusco A, Valle D, Tacchino RM. Growth
Bougneres P, Lebouc Y, Froguel P, Guy-Grand B. A mutation in the hormone secretion and leptin in morbid obesity before and after bil-
human leptin receptor gene causes obesity and pituitary dysfunction. iopancreatic diversion: Relationships with insulin and body composi-
Nature 392: 398-401, 1998. tion. J Clin Endocrinol Metab 89: 174-180, 2004.
96. Cocchi D, Colonna VD, Bagnasco M, Bonacci D, Muller EE. Leptin 120. de Zegher F, Devlieger H, Veldhuis JD. Properties of growth hormone
regulates GH secretion in the rat by acting on GHRH and somatostatin- and prolactin hypersecretion by the human infant on the day of birth.
ergic functions. J Endocrinol 162: 95-99, 1999. J Clin Endocrinol Metab 76: 1177-1181, 1993.
97. Coiro V, Volpi R, Bertoni P, Finzi G, Marcato A, Caiazza A, Colla R, 121. de Zegher F, Pernasetti F, Vanhole C, Devlieger H, Van den Berghe
Giacalone G, Rossi G, Chiodera P. Effect of potentiation of cholinergic G, Martial JA. The prenatal role of thyroid hormone evidenced by
tone by pyridostigmine on the GH response to GHRH in elderly men. fetomaternal Pit-1 deficiency. J Clin Endocrinol Metab 80: 3127-3130,
Gerontology 38: 217-222, 1992. 1995.
98. Colman RJ, Beasley TM, Kemnitz JW, Johnson SC, Weindruch R, 122. de Zegher F, Vanderschueren-Lodeweyckx M, Spitz B, Faijerson Y,
Anderson RM. Caloric restriction reduces age-related and all-cause Blomberg F, Beckers A, Hennen G, Frankenne F. Perinatal growth
mortality in rhesus monkeys. Nat Commun 5: 3557, 2014. hormone (GH) physiology: Effect of GH-releasing factor on maternal
99. Colmers WF, Bleakman D. Effects of neuropeptide Y on the electrical and fetal secretion of pituitary and placental GH. J Clin Endocrinol
properties of neurons. Trends Neurosci 17: 373-379, 1994. Metab 71: 520-522, 1990.
100. Conley LK, Teik JA, Deghenghi R, Imbimbo BP, Giustina A, Locatelli 123. Deghenghi R, Cananzi MM, Torsello A, Battisti C, Muller EE, Locatelli
V, Wehrenberg WB. Mechanism of action of hexarelin and GHRP- V. GH-releasing activity of Hexarelin, a new growth hormone releasing
6: Analysis of the involvement of GHRH and somatostatin in the rat. peptide, in infant and adult rats. Life Sci 54: 1321-1328, 1994.
Neuroendocrinology 61: 44-50, 1995. 124. Dekkers OM, Biermasz NR, Pereira AM, Romijn JA, Vandenbroucke
101. Conover CA, Boldt HB, Bale LK, Clifton KB, Grell JA, Mader JP. Mortality in acromegaly: A metaanalysis. J Clin Endocrinol Metab
JR, Mason EJ, Powell DR. Pregnancy-associated plasma protein-A2 93: 61-67, 2008.
(PAPP-A2): Tissue expression and biological consequences of gene 125. Dell’agnello C, Leo S, Agostino A, Szabadkai G, Tiveron C, Zulian
knockout in mice. Endocrinology 152: 2837-2844, 2011. A, Prelle A, Roubertoux P, Rizzuto R, Zeviani M. Increased longevity
102. Cook CB, Nippoldt TB, Kletter GB, Kelch RP, Marshall JC. Naloxone and refractoriness to Ca(2+)-dependent neurodegeneration in Surf1
increases the frequency of pulsatile luteinizing hormone secretion in knockout mice. Hum Mol Genet 16: 431-444, 2007.
women with hyperprolactinemia. J Clin Endocrinol Metab 73: 1099- 126. Demange L, Boeglin D, Moulin A, Mousseaux D, Ryan J, Bergé G,
1105, 1991. Gagne D, Heitz A, Perrissoud D, Locatelli V, Torsello A, Galleyrand
103. Copinschi G, Leproult R, Van Onderbergen A, Caufriez A, Cole KY, J-C, Fehrentz J-A, Martinez J. Synthesis and pharmacological in vitro
Schilling LM, Mendel CM, De Lepeleire I, Bolognese JA, Van Cauter and in vivo evaluations of novel triazole derivatives as ligands of the
E. Prolonged oral treatment with MK-677, a novel growth hormone ghrelin receptor. 1. J Med Chem 50: 1939-1957, 2007.
secretagogue, improves sleep quality in man. Neuroendocrinology 66: 127. Devesa J, Arce V, Lois N, Tresguerres JA, Lima L. Alpha 2-adrenergic
278-286, 1997. agonism enhances the growth hormone (GH) response to GH-releasing
104. Copinschi G, Nedeltcheva A, Leproult R, Morselli LL, Spiegel K, hormone through an inhibition of hypothalamic somatostatin release in
Martino E, Legros JJ, Weiss RE, Mockel J, Van Cauter E. Sleep normal men. J Clin Endocrinol Metab 71: 1581-1588, 1990.
disturbances, daytime sleepiness, and quality of life in adults with 128. Drouva SV, Epelbaum J, Tapia-Arancibia L, Laplante E, Kordon C.
growth hormone deficiency. J Clin Endocrinol Metab 95: 2195-2202, Opiate receptors modulate LHRH and SRIF release from mediobasal
2010. hypothalamic neurons. Neuroendocrinology 32: 163-167, 1981.
105. Cordido F, Peino R, Penalva A, Alvarez CV, Casanueva FF, Dieguez 129. Duhault J, Boulanger M, Chamorro S, Boutin JA, Della Zuana O,
C. Impaired growth hormone secretion in obese subjects is partially Douillet E, Fauchere JL, Feletou M, Germain M, Husson B, Vega AM,
reversed by acipimox-mediated plasma free fatty acid depression. J Clin Renard P, Tisserand F. Food intake regulation in rodents: Y5 or Y1
Endocrinol Metab 81: 914-918, 1996. NPY receptors or both? Can J Physiol Pharmacol 78: 173-185, 2000.
106. Cornford AS, Barkan AL, Hinko A, Horowitz JF. Suppression in growth 130. Durand D, Martin JB, Brazeau P. Evidence for a role of alpha-
hormone during overeating ameliorates the increase in insulin resistance adrenergic mechanisms in regulation of episodic growth hormone secre-
and cardiovascular disease risk. Am J Physiol Endocrinol Metab 303: tion in the rat. Endocrinology 100: 722-728, 1977.
E1264-1272, 2012. 131. Elster AD, Sanders TG, Vines FS, Chen MY. Size and shape of the
107. Cornford AS, Barkan AL, Horowitz JF. Rapid suppression of growth pituitary gland during pregnancy and post partum: Measurement with
hormone concentration by overeating: Potential mediation by hyperin- MR imaging. Radiology 181: 531-535, 1991.
sulinemia. J Clin Endocrinol Metab 96: 824-830, 2011. 132. English DE, Russell SM, Katz LS, Nicoll CS. Evidence for a role of
108. Corpas E, Harman SM, Blackman MR. Human growth hormone and the liver in the mammotrophic action of prolactin. Endocrinology 126:
human aging. Endocr Rev 14: 20-39, 1993. 2252-2256, 1990.
109. Coxam V, Davicco MJ, Dardillat C, Robelin J, Lefaivre J, Opmeer F, 133. Epelbaum J, Arancibia LT, Herman JP, Kordon C, Palkovits M. Topog-
Barlet JP. Regulation of growth hormone release in fetal calves. Biol raphy of median eminence somatostatinergic innervation. Brain Res
Neonate 54: 160-168, 1988. 230: 412-416, 1981.
110. Cummings DE, Merriam GR. Growth hormone therapy in adults. Annu 134. Eriksson L, Frankenne F, Eden S, Hennen G, Von Schoultz B. Growth
Rev Med 54: 513-533, 2003. hormone 24-h serum profiles during pregnancy–lack of pulsatility for
111. Curran AJ, Peacey SR, Shalet SM. Is maternal growth hormone essen- the secretion of the placental variant. Br J Obstet Gynaecol 96: 949-953,
tial for a normal pregnancy? Eur J Endocrinol 139: 54-58, 1998. 1989.
112. Daikoku S, Chikamori-Aoyama M, Tokuzen M, Okamura Y, Kagotani 135. Esposito M, Pellinen J, Kapas L, Szentirmai E. Impaired wake-
Y. Development of hypothalamic neurons in intraventricular grafts: promoting mechanisms in ghrelin receptor-deficient mice. Eur J Neu-
Expression of specific transmitter phenotypes. Dev Biol 126: 382-393, rosci 35: 233-243, 2012.
1988. 136. Ettigi P, Lal S, Martin JB, Friesen HG. Effectof sex, oral contracep-
113. Danger JM, Tonon MC, Jenks BG, Saint-Pierre S, Martel JC, Fasolo tives, and glucose loading on apomorphine-induced growth hormone
A, Breton B, Quirion R, Pelletier G, Vaudry H. Neuropeptide Y: Local- secretion. J Clin Endocrinol Metab 40: 1094-1098, 1975.
ization in the central nervous system and neuroendocrine functions. 137. Fagin JA, Fernandez-Mejia C, Melmed S. Pituitary insulin-like growth
Fundam Clin Pharmacol 4: 307-340, 1990. factor-I gene expression: Regulation by triiodothyronine and growth
114. Davidson MB. Effect of growth hormone on carbohydrate and lipid hormone. Endocrinology 125: 2385-2391, 1989.
metabolism. Endocr Rev 8: 115-131, 1987. 138. Fairhall KM, Mynett A, Robinson IC. Central effects of growth
115. Davies RR, Turner S, Johnston DG. Oral glucose inhibits growth hor- hormone-releasing hexapeptide (GHRP-6) on growth hormone release
mone secretion induced by human pancreatic growth hormone releasing are inhibited by central somatostatin action. J Endocrinol 144: 555-560,
factor 1-44 in normal man. Clin Endocrinol (Oxf) 21: 477-481, 1984. 1995.
116. de Gennaro Colonna V, Cattaneo E, Cocchi D, Muller EE, Maggi 139. Fairhall KM, Mynett A, Smith RG, Robinson IC. Consistent GH
A. Growth hormone regulation of growth hormone-releasing hormone responses to repeated injection of GH-releasing hexapeptide (GHRP-6)
gene expression. Peptides 9: 985-988, 1988. and the non-peptide GH secretagogue, L-692,585. J Endocrinol 145:
117. de Gennaro Colonna V, Fidone F, Cocchi D, Muller EE. Feedback 417-426, 1995.
effects of growth hormone on growth hormone-releasing hormone and 140. Faria AC, Bekenstein LW, Booth RA, Jr., Vaccaro VA, Asplin CM,
somatostatin are not evident in aged rats. Neurobiol Aging 14: 503-507, Veldhuis JD, Thorner MO, Evans WS. Pulsatile growth hormone release
1993. in normal women during the menstrual cycle. Clin Endocrinol (Oxf)
118. de los Frailes MT, Cacicedo L, Fernandez G, Tolon RM, Jesus Lorenzo 36: 591-596, 1992.
M, Aguado F, Sanchez Franco F. Role of locally produced growth 141. Feng DD, Yang SK, Loudes C, Simon A, Al-Sarraf T, Culler M, Alvear-
hormone-releasing factor in somatostatin regulation by fetal rat brain Perez R, Llorens-Cortes C, Chen C, Epelbaum J, Gardette R. Ghrelin
cells in culture. Neuroendocrinology 55: 221-229, 1992. and obestatin modulate growth hormone-releasing hormone release and

724 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

synaptic inputs onto growth hormone-releasing hormone neurons. Eur 165. Gao P, Ishige A, Murakami Y, Nakata H, Oka J, Munakata K,
J Neurosci 34: 732-744, 2011. Yamamoto M, Nishimura K, Watanabe K. Maternal stress affects post-
142. Fetissov SO, Kopp J, Hokfelt T. Distribution of NPY receptors in the natal growth and the pituitary expression of prolactin in mouse off-
hypothalamus. Neuropeptides 38: 175-188, 2004. spring. J Neurosci Res 89: 329-340, 2011.
143. Flint DJ, Knight CH. Interactions of prolactin and growth hormone 166. Garrel DR, Bajard L, Harfouche M, Tourniaire J. Effect of sustained
(GH) in the regulation of mammary gland function and epithelial cell hyperglycemia on GHRH induced GH secretion in man. Diabete Metab
survival. J Mammary Gland Biol Neoplasia 2: 41-48, 1997. 15: 251-254, 1989.
144. Flint DJ, Vernon RG. Effects of food restriction on the responses of the 167. Gatford KL, Boyce JM, Blackmore K, Smits RJ, Campbell RG, Owens
mammary gland and adipose tissue to prolactin and growth hormone in PC. Long-term, but not short-term, treatment with somatotropin during
the lactating rat. J Endocrinol 156: 299-305, 1998. pregnancy in underfed pigs increases the body size of progeny at birth.
145. Fodor M, Csaba Z, Epelbaum J, Vaudry H, Jegou S. Interrelations J Anim Sci 82: 93-101, 2004.
between hypothalamic somatostatin and proopiomelanocortin neurons. 168. Gatford KL, Ekert JE, Blackmore K, De Blasio MJ, Boyce JM, Owens
J Neuroendocrinol 10: 75-78, 1998. JA, Campbell RG, Owens PC. Variable maternal nutrition and growth
146. Fodor M, Csaba Z, Kordon C, Epelbaum J. Growth hormone-releasing hormone treatment in the second quarter of pregnancy in pigs alter
hormone, somatostatin, galanin and beta-endorphin afferents to the semitendinosus muscle in adolescent progeny. Br J Nutr 90: 283-293,
hypothalamic periventricular nucleus. J Chem Neuroanat 8: 61-73, 2003.
1994. 169. Ghigo E, Ceda GP, Valcavi R, Goffi S, Zini M, Mucci M, Valenti
147. Fodor M, Slama A, Guillaume V, Videau C, Csaba Z, Oliver C, Epel- G, Muller EE, Camanni F. Effect of 15-day treatment with growth-
baum J. Distribution and pharmacological characterization of somato- hormone-releasing hormone alone or combined with different doses of
statin receptor binding sites in the sheep brain. J Chem Neuroanat 12: arginine on the reduced somatotrope responsiveness to the neurohor-
175-182, 1997. mone in normal aging. Eur J Endocrinol 132: 32-36, 1995.
148. Folli F, Ghidella S, Bonfanti L, Kahn CR, Merighi A. The early intra- 170. Giordano R, Bonelli L, Marinazzo E, Ghigo E, Arvat E. Growth
cellular signaling pathway for the insulin/insulin-like growth factor hormone treatment in human ageing: Benefits and risks. Hormones
receptor family in the mammalian central nervous system. Mol Neuro- (Athens) 7: 133-139, 2008.
biol 13: 155-183, 1996. 171. Girod JP, Brotman DJ. The metabolic syndrome as a vicious cycle:
149. Franco C, Brandberg J, Lonn L, Andersson B, Bengtsson BA, Johanns- Does obesity beget obesity? Med Hypotheses 60: 584-589, 2003.
son G. Growth hormone treatment reduces abdominal visceral fat 172. Giustina A, Bresciani E, Tassi C, Girelli A, Valentini U. Effect of
in postmenopausal women with abdominal obesity: A 12-month pyridostigmine on the growth hormone response to growth hormone-
placebo-controlled trial. J Clin Endocrinol Metab 90: 1466-1474, releasing hormone in lean and obese type II Diabetic patients.
2005. Metabolism 43: 893-898, 1994.
150. Frankenne F, Closset J, Gomez F, Scippo ML, Smal J, Hennen G. The 173. Giustina A, Scalvini T, Tassi C, Desenzani P, Poiesi C, Wehrenberg
physiology of growth hormones (GHs) in pregnant women and partial WB, Rogol AD, Veldhuis JD. Maturation of the regulation of growth
characterization of the placental GH variant. J Clin Endocrinol Metab hormone secretion in young males with hypogonadotropic hypogo-
66: 1171-1180, 1988. nadism pharmacologically exposed to progressive increments in serum
151. Frantz AG, Rabkin MT. Effects of estrogen and sex difference on testosterone. J Clin Endocrinol Metab 82: 1210-1219, 1997.
secretion of human growth hormone. J Clin Endocrinol Metab 25: 174. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of
1470-1480, 1965. growth hormone secretion in experimental animals and the human.
152. Fraser RA, Siminoski K, Harvey S. Growth hormone receptor gene: Endocr Rev 19: 717-797, 1998.
Novel expression in pituitary tissue. J Endocrinol 128: R9-11, 1991. 175. Glenn KC. Regulation of release of somatotropin from in vitro cultures
153. Frasier SD. A preview of growth hormone stimulation tests in children. of bovine and porcine pituitary cells. Endocrinology 118: 2450-2457,
Pediatrics 53: 929-937, 1974. 1986.
154. Frederich RC, Hamann A, Anderson S, Lollmann B, Lowell BB, Flier 176. Gluckman PD, Gunn AJ, Wray A, Cutfield WS, Chatelain PG, Guilbaud
JS. Leptin levels reflect body lipid content in mice-evidence for diet O, Ambler GR, Wilton P, Albertsson-Wikland K. Congenital idiopathic
induced resistance to leptin action. Nat Med 1: 1311-1314, 1995. growth hormone deficiency associated with prenatal and early postnatal
155. Frieboes RM, Antonijevic IA, Held K, Murck H, Pollmacher T, Uhr growth failure. The International Board of the Kabi Pharmacia Interna-
M, Steiger A. Hexarelin decreases slow-wave sleep and stimulates the tional Growth Study. J Pediatr 121: 920-923, 1992.
secretion of GH, ACTH, cortisol and prolactin during sleep in healthy 177. Gluckman PD, Pinal CS. Maternal-placental-fetal interactions in the
volunteers. Psychoneuroendocrinology 29: 851-860, 2004. endocrine regulation of fetal growth: Role of somatotrophic axes.
156. Frieboes RM, Murck H, Antonijevic IA, Steiger A. Effects of growth Endocrine 19: 81-89, 2002.
hormone-releasing peptide-6 on the nocturnal secretion of GH, ACTH 178. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P,
and cortisol and on the sleep EEG in man: Role of routes of adminis- Bhattacharya S, Carpenter R, Grossman AB, Korbonits M. The tissue
tration. J Neuroendocrinol 11: 473-478, 1999. distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-
157. Frieboes RM, Murck H, Maier P, Schier T, Holsboer F, Steiger A. R, in humans. J Clin Endocrinol Metab 87: 2988, 2002.
Growth hormone-releasing peptide-6 stimulates sleep, growth hor- 179. Golstein J, Van Cauter E, Desir D, Noel P, Spire JP, Refetoff S, Copin-
mone, ACTH and cortisol release in normal man. Neuroendocrinology schi G. Effects of “jet lag” on hormonal patterns. IV. Time shifts increase
61: 584-589, 1995. growth hormone release. J Clin Endocrinol Metab 56: 433-440, 1983.
158. Frisch RE, Revelle R. The height and weight of girls and boys at the 180. Gong Z, Kennedy O, Sun H, Wu Y, Williams GA, Klein L, Cardoso
time of initiation of the adolescent growth spurt in height and weight L, Matheny RW, Jr, Hubbard GB, Ikeno Y, Farrar RP, Schaffler MB,
and the relationship to menarche. Hum Biol 43: 140-159, 1971. Adamo ML, Muzumdar RH, Yakar S. Reductions in serum IGF-1 dur-
159. Frohman LA, Downs TR, Clarke IJ, Thomas GB. Measurement of ing aging impair health span. Aging cell 13: 408-418, 2014.
growth hormone-releasing hormone and somatostatin in hypothalamic- 181. Guan XM, Yu H, Palyha OC, McKee KK, Feighner SD, Sirinaths-
portal plasma of unanesthetized sheep. Spontaneous secretion and inghji DJ, Smith RG, Van der Ploeg LH, Howard AD. Distribution of
response to insulin-induced hypoglycemia. J Clin Invest 86: 17-24, mRNA encoding the growth hormone secretagogue receptor in brain
1990. and peripheral tissues. Brain Res Mol Brain Res 48: 23-29, 1997.
160. Frohman LA, Kineman RD. Growth hormone-releasing hormone: Dis- 182. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M,
covery, regulation and actions. In: Kostyo JL, editor. Handbook of Phys- Madia F, Cheng CW, Hwang D, Martin-Montalvo A, Saavedra J, Ingles
iology. New York, Oxford: Oxford University Press for the American S, de Cabo R, Cohen P, Longo VD. Growth hormone receptor deficiency
Physiology Society, 1999, pp. 187-219. is associated with a major reduction in pro-aging signaling, cancer, and
161. Gabriel SM, Rivkin A, Mercado J, Jr. The galanin antagonist, M- diabetes in humans. Sci Transl Med 3: 70ra13, 2011.
15, inhibits growth hormone release in rats. Peptides 14: 633-636, 183. Guillaume V, Magnan E, Cataldi M, Dutour A, Sauze N, Renard M,
1993. Razafindraibe H, Conte-Devolx B, Deghenghi R, Lenaerts V. Growth
162. Gahete MD, Cordoba-Chacon J, Anadumaka CV, Lin Q, Bruning hormone (GH)-releasing hormone secretion is stimulated by a new
JC, Kahn CR, Luque RM, Kineman RD. Elevated GH/IGF-I, due to GH-releasing hexapeptide in sheep. Endocrinology 135: 1073-1076,
somatotrope-specific loss of both IGF-I and insulin receptors, alters 1994.
glucose homeostasis and insulin sensitivity in a diet-dependent man- 184. Gunn AJ, Gunn TR, Rabone DL, Breier BH, Blum WF, Gluckman PD.
ner. Endocrinology 152: 4825-4837, 2011. Growth hormone increases breast milk volumes in mothers of preterm
163. Gahete MD, Cordoba-Chacon J, Lin Q, Bruning JC, Kahn CR, Castano infants. Pediatrics 98: 279-282, 1996.
JP, Christian H, Luque RM, Kineman RD. Insulin and IGF-I inhibit 185. Gutierrez JA, Solenberg PJ, Perkins DR, Willency JA, Knierman MD,
GH synthesis and release in vitro and in vivo by separate mechanisms. Jin Z, Witcher DR, Luo S, Onyia JE, Hale JE. Ghrelin octanoylation
Endocrinology 154: 2410-2420, 2013. mediated by an orphan lipid transferase. Proc Natl Acad Sci U S A 105:
164. Ganzetti I, De Gennaro V, Redaelli M, Muller EE, Cocchi D. Effect of 6320-6325, 2008.
hypophysectomy and growth hormone replacement on hypothalamic 186. Hadsell DL, Parlow AF, Torres D, George J, Olea W. Enhancement of
GHRH. Peptides 7: 1011-1014, 1986. maternal lactation performance during prolonged lactation in the mouse

Volume 6, April 2016 725


Growth Hormone Axis Comprehensive Physiology

by mouse GH and long-R3-IGF-I is linked to changes in mammary 211. Holloway L, Butterfield G, Hintz RL, Gesundheit N, Marcus R. Effects
signaling and gene expression. J Endocrinol 198: 61-70, 2008. of recombinant human growth hormone on metabolic indices, body
187. Haggi ES, Torres AI, Maldonado CA, Aoki A. Regression of redun- composition, and bone turnover in healthy elderly women. J Clin
dant lactotrophs in rat pituitary gland after cessation of lactation. Endocrinol Metab 79: 470-479, 1994.
J Endocrinol 111: 367-373, 1986. 212. Holst B, Brandt E, Bach A, Heding A, Schwartz TW. Nonpeptide and
188. Hajdu I, Obal F, Jr., Fang J, Krueger JM, Rollo CD. Sleep of transgenic peptide growth hormone secretagogues act both as ghrelin receptor
mice producing excess rat growth hormone. Am J Physiol Regul Integr agonist and as positive or negative allosteric modulators of ghrelin
Comp Physiol 282: R70-76, 2002. signaling. Mol Endocrinol 19: 2400-2411, 2005.
189. Halem HA, Taylor JE, Dong JZ, Shen Y, Datta R, Abizaid A, Diano 213. Holst B, Cygankiewicz A, Jensen TH, Ankersen M, Schwartz TW.
S, Horvath TL, Culler MD. A novel growth hormone secretagogue- High constitutive signaling of the ghrelin receptor–identification of a
1a receptor antagonist that blocks ghrelin-induced growth hormone potent inverse agonist. Mol Endocrinol 17: 2201-2210, 2003.
secretion but induces increased body weight gain. Neuroendocrinology 214. Holst B, Egerod KL, Schild E, Vickers SP, Cheetham S, Gerlach L-
81: 339-349, 2005. O, Storjohann L, Stidsen CE, Jones R, Beck-Sickinger AG, Schwartz
190. Hall K, Enberg G, Hellem E, Lundin G, Ottosson-Seeberger A, Sara TW. GPR39 signaling is stimulated by zinc ions but not by obestatin.
V, Trygstad O, Ofverholm U. Somatomedin levels in pregnancy: Lon- Endocrinology 148: 13-20, 2007.
gitudinal study in healthy subjects and patients with growth hormone 215. Holst B, Holliday ND, Bach A, Elling CE, Cox HM, Schwartz TW.
deficiency. J Clin Endocrinol Metab 59: 587-594, 1984. Common structural basis for constitutive activity of the ghrelin receptor
191. Hansen AP, Johansen K. Diurnal patterns of blood glucose, serum family. J Biol Chem 279: 53806-53817, 2004.
free fatty acids, insulin, glucagon and growth hormone in normals and 216. Holst B, Schwartz TW. Ghrelin receptor mutations–too little height and
juvenile diabetics. Diabetologia 6: 27-33, 1970. too much hunger. J Clin Invest 116: 637-641, 2006.
192. Harfstrand A, Eneroth P, Agnati L, Fuxe K. Further studies on the 217. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC,
effects of central administration of neuropeptide Y on neuroendocrine Cervera P, Le Bouc Y. IGF-1 receptor regulates lifespan and resistance
function in the male rat: Relationship to hypothalamic catecholamines. to oxidative stress in mice. Nature 421: 182-187, 2003.
Regul Pept 17: 167-179, 1987. 218. Horner JM, Liu F, Hintz RL. Comparison of [125I]somatomedin A and
193. Hartman ML, Veldhuis JD, Johnson ML, Lee MM, Alberti KG, Samo- [125I]somatomedin C radioreceptor assays for somatomedin peptide
jlik E, Thorner MO. Augmented growth hormone (GH) secretory burst content in whole and acid-chromatographed plasma. J Clin Endocrinol
frequency and amplitude mediate enhanced GH secretion during a two- Metab 47: 1287-1295, 1978.
day fast in normal men. J Clin Endocrinol Metab 74: 757-765, 1992. 219. Horvath S, Palkovits M, Gorcs T, Arimura A. Electron microscopic
194. Hassouna R, Labarthe A, Zizzari P, Videau C, Culler M, Epelbaum J, immunocytochemical evidence for the existence of bidirectional synap-
Tolle V. Actions of Agonists and Antagonists of the ghrelin/GHS- tic connections between growth hormone-releasing hormone- and
R Pathway on GH Secretion, Appetite, and cFos Activity. Front somatostatin-containing neurons in the hypothalamus of the rat. Brain
Endocrinol (Lausanne) 4: 25, 2013. Res 481: 8-15, 1989.
195. Hassouna R, Zizzari P, Tomasetto C, Veldhuis JD, Fiquet O, Labarthe 220. Horvath TL, Kalra SP, Naftolin F, Leranth C. Morphological evidence
A, Cognet J, Steyn F, Chen C, Epelbaum J, Tolle V. An early reduction for a galanin-opiate interaction in the rat mediobasal hypothalamus.
in GH peak amplitude in preproghrelin-deficient male mice has a minor J Neuroendocrinol 7: 579-588, 1995.
impact on linear growth. Endocrinology 155: 3561-3571, 2014. 221. Horvath TL, Naftolin F, Kalra SP, Leranth C. Neuropeptide-Y innerva-
196. Hassouna R, Zizzari P, Viltart O, Yang SK, Gardette R, Videau C, tion of beta-endorphin-containing cells in the rat mediobasal hypothala-
Badoer E, Epelbaum J, Tolle V. A natural variant of obestatin, Q90L, mus: A light and electron microscopic double immunostaining analysis.
inhibits ghrelin’s action on food intake and GH secretion and targets Endocrinology 131: 2461-2467, 1992.
NPY and GHRH neurons in mice. PLoS One 7: e51135, 2012. 222. Horvath TL, Naftolin F, Leranth C. GABAergic and catecholaminergic
197. Hayford JT, Danney MM, Hendrix JA, Thompson RG. Integrated con- innervation of mediobasal hypothalamic beta-endorphin cells project-
centration of growth hormone in juvenile-onset diabetes. Diabetes 29: ing to the medial preoptic area. Neuroscience 51: 391-399, 1992.
391-398, 1980. 223. Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosen-
198. He C, Tsend-Ayush E, Myers MA, Forbes BE, Grutzner F. Changes in blum CI, Hamelin M, Hreniuk DL, Palyha OC, Anderson J, Paress PS,
the ghrelin hormone pathway maybe part of an unusual gastric system Diaz C, Chou M, Liu KK, McKee KK, Pong SS, Chaung LY, Elbrecht
in monotremes. Gen Comp Endocrinol 191: 74-82, 2013. A, Dashkevicz M, Heavens R, Rigby M, Sirinathsinghji DJ, Dean DC,
199. Herzog H. Neuropeptide Y and energy homeostasis: Insights from Y Melillo DG, Patchett AA, Nargund R, Griffin PR, DeMartino JA, Gupta
receptor knockout models. Eur J Pharmacol 480: 21-29, 2003. SK, Schaeffer JM, Smith RG, Van der Ploeg LH. A receptor in pituitary
200. Higuchi T, Takahashi Y, Takahashi K, Niimi Y, Miyasita A. Twenty- and hypothalamus that functions in growth hormone release. Science
four-hour secretory patterns of growth hormone, prolactin, and cortisol 273: 974-977, 1996.
in narcolepsy. J Clin Endocrinol Metab 49: 197-204, 1979. 224. Huang L, Steyn FJ, Tan HY, Xie TY, Veldhuis JD, Ngo ST, Chen
201. Hill JM, Lesniak MA, Pert CB, Roth J. Autoradiographic localization C. The decline in pulsatile GH secretion throughout early adulthood
of insulin receptors in rat brain: Prominence in olfactory and limbic in mice is exacerbated by dietary-induced weight gain. Endocrinology
areas. Neuroscience 17: 1127-1138, 1986. 153: 4380-4388, 2012.
202. Hills FA, English J, Chard T. Circulating levels of IGF-I and IGF- 225. Huang L, Tan HY, Fogarty MJ, Andrews ZB, Veldhuis JD, Herzog
binding protein-1 throughout pregnancy: Relation to birthweight and H, Steyn FJ, Chen C. Actions of NPY, and Its Y1 and Y2 receptors
maternal weight. J Endocrinol 148: 303-309, 1996. on pulsatile growth hormone secretion during the fed and fasted state.
203. Hindmarsh P, Brook C, Rodeck C, Nicolaides K. Hormonal levels in J Neurosci 34: 16309-16319, 2014.
the human fetus between 14 and 22 weeks gestation. Early human 226. Hulting AL, Meister B, Carlsson L, Hilding A, Isaksson O. On the role
development 15: 253-254, 1987. of the peptide galanin in regulation of growth hormone secretion. Acta
204. Hisano S, Tsuruo Y, Kagotani Y, Daikoku S, Chihara K. Immunohis- endocrinologica 125: 518-525, 1991.
tochemical evidence for synaptic connections between neuropeptide 227. Hunter WM, Willoughby JM, Strong JA. Plasma insulin and growth
Y-containing axons and periventricular somatostatin neurons in the hormone during 22-hour fasts and after graded glucose loads in six
anterior hypothalamus in rats. Brain Res 520: 170-177, 1990. healthy adults. J Endocrinol 40: 297-311, 1968.
205. Ho KY, Veldhuis JD, Johnson ML, Furlanetto R, Evans WS, Alberti K, 228. Ilias I, Ghizzoni L, Mastorakos G. Orderliness of cortisol, growth hor-
Thorner MO. Fasting enhances growth-hormone secretion and ampli- mone, and leptin secretion in short-normal pre-pubertal boys and girls.
fies the complex rhythms of growth-hormone secretion in man. J Clin Med Sci Monit 15: CR242-247, 2009.
Invest 81: 968-975, 1988. 229. Imura H, Nakai Y, Yoshimi T. Effect of 5-hydroxytryptophan (5-HTP)
206. Hoffman DL, Baker BL. Effect of treatment with growth hormone on on growth hormone and ACTH release in man. J Clin Endocrinol Metab
somatostatin in the median eminence of hypophysectomized rats. Proc 36: 204-206, 1973.
Soc Exp Biol Med 156: 265-271, 1977. 230. Inhoff T, Mönnikes H, Noetzel S, Stengel A, Goebel M, Dinh QT,
207. Hokfelt T, Meister B, Villar MJ, Ceccatelli S, Corts R, Schalling M, Riedl A, Bannert N, Wisser A-S, Wiedenmann B, Klapp BF, Taché Y,
Everitt B. Colocalization of messenger substances with special refer- Kobelt P. Desacyl ghrelin inhibits the orexigenic effect of peripherally
ence to the hypothalamic arcuate and paraventricular nuclei. Prog Clin injected ghrelin in rats. Peptides 29: 2159-2168, 2008.
Biol Res 342: 257-264, 1990. 231. Inhoff T, Wiedenmann B, Klapp BF, Mönnikes H, Kobelt P. Is desacyl
208. Hokfelt T, Tatemoto K. Galanin: A multitalented neuropeptide. EXS ghrelin a modulator of food intake? Peptides 30: 991-994, 2009.
102: 1-5, 2010. 232. Iqbal J, Manley TR, Ciofi P, Clarke IJ. Reduction in adiposity affects the
209. Holdaway IM, Bolland MJ, Gamble GD. A meta-analysis of the effect extent of afferent projections to growth hormone-releasing hormone and
of lowering serum levels of GH and IGF-I on mortality in acromegaly. somatostatin neurons and the degree of colocalization of neuropeptides
Eur J Endocrinol 159: 89-95, 2008. in growth hormone-releasing hormone and somatostatin cells of the
210. Holl RW, Hartman ML, Veldhuis JD, Taylor WM, Thorner MO. Thirty- ovine hypothalamus. Endocrinology 146: 4776-4785, 2005.
second sampling of plasma growth hormone in man: Correlation with 233. Iranmanesh A, Lizarralde G, Veldhuis JD. Age and relative adiposity
sleep stages. J Clin Endocrinol Metab 72: 854-861, 1991. are specific negative determinants of the frequency and amplitude of

726 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

growth hormone (GH) secretory bursts and the half-life of endogenous 257. Keen-Rhinehart E, Bartness TJ. NPY Y1 receptor is involved in ghrelin-
GH in healthy men. J Clin Endocrinol Metab 73: 1081-1088, 1991. and fasting-induced increases in foraging, food hoarding, and food
234. Ismail IS, Scanlon MF, Peters JR. Cholinergic control of growth hor- intake. Am J Physiol Regul Integr Comp Physiol 292: R1728-1737,
mone (GH) responses to GH-releasing hormone in insulin dependent 2007.
diabetics: Evidence for attenuated hypothalamic somatostatinergic tone 258. Kelijman M, Frohman LA. Beta-adrenergic modulation of growth
and decreased GH autofeedback. Clin Endocrinol (Oxf) 38: 149-157, hormone (GH) autofeedback on sleep-associated and pharmacologi-
1993. cally induced GH secretion. J Clin Endocrinol Metab 69: 1187-1194,
235. Ismailogullari S, Tanriverdi F, Kelestimur F, Aksu M. Sleep architecture 1989.
in Sheehan’s syndrome before and 6 months after growth hormone 259. Kelley DE, Wing R, Buonocore C, Sturis J, Polonsky K, Fitzsimmons
replacement therapy. Psychoneuroendocrinology 34: 212-219, 2009. M. Relative effects of calorie restriction and weight loss in noninsulin-
236. Iwakura H, Ariyasu H, Li Y, Kanamoto N, Bando M, Yamada G, dependent diabetes mellitus. J Clin Endocrinol Metab 77: 1287-1293,
Hosoda H, Hosoda K, Shimatsu A, Nakao K, Kangawa K, Akamizu 1993.
T. A mouse model of ghrelinoma exhibited activated growth hormone- 260. Kelly JM, Bianchi MT. Mammalian sleep genetics. Neurogenetics 13:
insulin-like growth factor I axis and glucose intolerance. Am J Physiol 287-326, 2012.
Endocrinol Metab 297: E802-811, 2009. 261. Kerkhofs M, Van Cauter E, Van Onderbergen A, Caufriez A, Thorner
237. Jaffe CA, Friberg RD, Barkan AL. Suppression of growth hormone MO, Copinschi G. Sleep-promoting effects of growth hormone-
(GH) secretion by a selective GH-releasing hormone (GHRH) antag- releasing hormone in normal men. Am J Physiol 264: E594-E598,
onist. Direct evidence for involvement of endogenous GHRH in the 1993.
generation of GH pulses. J Clin Invest 92: 695-701, 1993. 262. Khatib N, Gaidhane S, Gaidhane AM, Khatib M, Simkhada P, Gode
238. Japon MA, Rubinstein M, Low MJ. In situ hybridization analysis of D, Zahiruddin QS. Ghrelin: Ghrelin as a regulatory Peptide in growth
anterior pituitary hormone gene expression during fetal mouse devel- hormone secretion. J Clin Diagn Res 8: MC13-MC17, 2014.
opment. J Histochem Cytochem 42: 1117-1125, 1994. 263. Kineman RD, Gahete MD, Luque RM. Identification of a mouse ghrelin
239. Jordan V, Dieguez C, Lafaffian I, Rodriguez-Arnao MD, Gomez-Pan gene transcript that contains intron 2 and is regulated in the pituitary
A, Hall R, Scanlon MF. Influence of dopaminergic, adrenergic and and hypothalamus in response to metabolic stress. J Mol Endocrinol
cholinergic blockade and TRH administration on GH responses to GRF 38: 511-521, 2007.
1-29. Clin Endocrinol (Oxf) 24: 291-298, 1986. 264. Kirchner H, Gutierrez JA, Solenberg PJ, Pfluger PT, Czyzyk TA, Wil-
240. Jorgensen JO, Christiansen JS. Clinical aspects of growth hormone lency JA, Schürmann A, Joost H-G, Jandacek RJ, Hale JE, Heiman
deficiency in adults. An overview. Front Horm Res 33: 1-20, 2005. ML, Tschöp MH. GOAT links dietary lipids with the endocrine control
241. Kabayama Y, Kato Y, Murakami Y, Tanaka H, Imura H. Stimulation of energy balance. Nat Med 15: 741-745, 2009.
by alpha-adrenergic mechanisms of the secretion of growth hormone- 265. Kirsz K, Szczesna M, Dudek K, Bartlewski PM, Zieba DA. Influence
releasing factor (GRF) from perifused rat hypothalamus. Endocrinology of season and nutritional status on the direct effects of leptin, orexin-A
119: 432-434, 1986. and ghrelin on luteinizing hormone and growth hormone secretion in
242. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to the ovine pituitary explant model. Domest Anim Endocrinol 48: 69-76,
insulin resistance and type 2 diabetes. Nature 444: 840-846, 2006. 2014.
243. Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS. Interacting 266. Kishi T, Aschkenasi CJ, Choi BJ, Lopez ME, Lee CE, Liu H, Hollenberg
appetite-regulating pathways in the hypothalamic regulation of body AN, Friedman JM, Elmquist JK. Neuropeptide Y Y1 receptor mRNA
weight. Endocr Rev 20: 68-100, 1999. in rodent brain: Distribution and colocalization with melanocortin-4
244. Kamegai J, Minami S, Sugihara H, Hasegawa O, Higuchi H, Wak- receptor. J Comp Neurol 482: 217-243, 2005.
abayashi I. Growth hormone receptor gene is expressed in neuropeptide 267. Kitajima N, Chihara K, Abe H, Okimura Y, Shakutsui S. Galanin
Y neurons in hypothalamic arcuate nucleus of rats. Endocrinology 137: stimulates immunoreactive growth hormone-releasing factor secretion
2109-2112, 1996. from rat hypothalamic slices perifused in vitro. Life Sci 47: 2371-2376,
245. Kamegai J, Wakabayashi I, Sugihara H, Minami S, Kitamura T, Yamada 1990.
J. Growth hormone secretion in stalk-sectioned rats. Acta endocrino- 268. Kletzky OA, Rossman F, Bertolli SI, Platt LD, Mishell DR, Jr. Dynam-
logica 124: 700-706, 1991. ics of human chorionic gonadotropin, prolactin, and growth hormone
246. Kanatsuka A, Makino H, Matsushima Y, Osegawa M, Yamamoto M, in serum and amniotic fluid throughout normal human pregnancy. Am
Kumagai A. Effect of hypophysectomy and growth hormone admin- J Obstet Gynecol 151: 878-884, 1985.
istration on somatostatin content in the rat hypothalamus. Neuroen- 269. Kluge M, Gazea M, Schussler P, Genzel L, Dresler M, Kleyer S, Uhr
docrinology 29: 186-190, 1979. M, Yassouridis A, Steiger A. Ghrelin increases slow wave sleep and
247. Kansal PC, Buse J, Talbert OR, Buse MG. The effect of L-dopa on stage 2 sleep and decreases stage 1 sleep and REM sleep in elderly men
plasma growth hormone, insulin, and thyroxine. J Clin Endocrinol but does not affect sleep in elderly women. Psychoneuroendocrinology
Metab 34: 99-105, 1972. 35: 297-304, 2010.
248. Kappeler L, De Magalhaes Filho C, Dupont J, Leneuve P, Cervera 270. Kluge M, Schussler P, Bleninger P, Kleyer S, Uhr M, Weikel JC,
P, Perin L, Loudes C, Blaise A, Klein R, Epelbaum J, Le Bouc Y, Yassouridis A, Zuber V, Steiger A. Ghrelin alone or co-administered
Holzenberger M. Brain IGF-1 receptors control mammalian growth with GHRH or CRH increases non-REM sleep and decreases REM
and lifespan through a neuroendocrine mechanism. PLoS Biol 6: e254, sleep in young males. Psychoneuroendocrinology 33: 497-506, 2008.
2008. 271. Kluge M, Schussler P, Zuber V, Kleyer S, Yassouridis A, Dresler
249. Kar S, Chabot JG, Quirion R. Quantitative autoradiographic localiza- M, Uhr M, Steiger A. Ghrelin enhances the nocturnal secretion of
tion of [125I]insulin-like growth factor I, [125I]insulin-like growth cortisol and growth hormone in young females without influencing
factor II, and [125I]insulin receptor binding sites in developing and sleep. Psychoneuroendocrinology 32: 1079-1085, 2007.
adult rat brain. J Comp Neurol 333: 375-397, 1993. 272. Kluge M, Schussler P, Zuber V, Yassouridis A, Steiger A. Ghrelin
250. Karpe F, Dickmann JR, Frayn KN. Fatty acids, obesity, and insulin administered in the early morning increases secretion of cortisol and
resistance: Time for a reevaluation. Diabetes 60: 2441-2449, 2011. growth hormone without affecting sleep. Psychoneuroendocrinology
251. Katakami H, Arimura A, Frohman LA. Growth hormone (GH)- 32: 287-292, 2007.
releasing factor stimulates hypothalamic somatostatin release: An 273. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K.
inhibitory feedback effect on GH secretion. Endocrinology 118: 1872- Ghrelin is a growth-hormone-releasing acylated peptide from stomach.
1877, 1986. Nature 402: 656-660, 1999.
252. Katakami H, Arimura A, Frohman LA. Involvement of hypothalamic 274. Kojima M, Kangawa K. Ghrelin: Structure and function. Physiol Rev
somatostatin in the suppression of growth hormone secretion by central 85: 495-522, 2005.
corticotropin-releasing factor in conscious male rats. Neuroendocrinol- 275. Kokka N, Garcia JF, Morgan M, George R. Immunoassay of plasma
ogy 41: 390-393, 1985. growth hormone in cats following fasting and administration of insulin,
253. Katakami H, Downs TR, Frohman LA. Effect of hypophysectomy on arginine, 2-deoxyglucose and hypothalamic extract. Endocrinology 88:
hypothalamic growth hormone-releasing factor content and release in 359-366, 1971.
the rat. Endocrinology 120: 1079-1082, 1987. 276. Kolakowski LF, Jr., O’Neill GP, Howard AD, Broussard SR, Sulli-
254. Kato Y, Chihara K, Ogo S, Imura H. Effects of hypothalamic surgery van KA, Feighner SD, Sawzdargo M, Nguyen T, Kargman S, Shiao
and somatostatin on chlorpromazine-induced growth hormone release LL, Hreniuk DL, Tan CP, Evans J, Abramovitz M, Chateauneuf A,
in rats. Endocrinology 95: 1608-1613, 1974. Coulombe N, Ng G, Johnson MP, Tharian A, Khoshbouei H, George
255. Kato Y, Dupre J, Beck JC. Plasma growth hormone in the anes- SR, Smith RG, O’Dowd BF. Molecular characterization and expression
thetized rat. Effects of dibutyryl cyclic AMP, prostaglandin E 1, adren- of cloned human galanin receptors GALR2 and GALR3. J Neurochem
ergic agents, vasopressin, chlorpromazine, amphetamine and L-dopa. 71: 2239-2251, 1998.
Endocrinology 93: 135-146, 1973. 277. Kopchick JJ, List EO, Kelder B, Gosney ES, Berryman DE. Evaluation
256. Kato Y, Matsushita N, Katagami H, Imura H. The mechanism of sero- of growth hormone (GH) action in mice: Discovery of GH receptor
toninergic regulation of prolactin and growth hormone secretion in rats antagonists and clinical indications. Mol Cell Endocrinol 386: 34-45,
(author’s transl). Horumon To Rinsho 28: 627-632, 1980. 2014.

Volume 6, April 2016 727


Growth Hormone Axis Comprehensive Physiology

278. Kopelman PG, Mason AC, Noonan K, Monson JP. Growth hormone 300. Lima L, Arce V, Tresguerres JA, Devesa J. Studies on alpha 2-
response to growth hormone releasing factor in diabetic men. Clin adrenergic modulation of hypothalamic somatostatin secretion in rats.
Endocrinol (Oxf) 28: 33-38, 1988. Life Sci 53: 665-668, 1993.
279. Krieg RJ, Perkins SN, Johnson JH, Rogers JP, Arimura A, Cronin MJ. 301. Lin S, Lin EJ, Boey D, Lee NJ, Slack K, During MJ, Sainsbury A,
Beta-adrenergic stimulation of growth hormone (GH) release in vivo, Herzog H. Fasting inhibits the growth and reproductive axes via distinct
and subsequent inhibition of GH-releasing factor-induced GH secre- Y2 and Y4 receptor-mediated pathways. Endocrinology 148: 2056-
tion. Endocrinology 122: 531-537, 1988. 2065, 2007.
280. Krzisnik C, Grguric S, Cvijovic K, Laron Z. Longevity of the hypopi- 302. Lindstrom P, Savendahl L. Effects of galanin on growth hormone
tuitary patients from the island Krk: A follow-up study. Pediatr release in isolated cultured rat somatotrophs. Acta endocrinologica
Endocrinol Rev 7: 357-362, 2010. 129: 268-272, 1993.
281. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, 303. Link K, Blizzard RM, Evans WS, Kaiser DL, Parker MW, Rogol AD.
McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura The effect of androgens on the pulsatile release and the twenty-four-
I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M. Suppres- hour mean concentration of growth hormone in peripubertal males.
sion of aging in mice by the hormone Klotho. Science 309: 1829-1833, J Clin Endocrinol Metab 62: 159-164, 1986.
2005. 304. Liposits Z, Merchenthaler I, Paull WK, Flerko B. Synaptic communi-
282. Labarthe A, Fiquet O, Hassouna R, Zizzari P, Lanfumey L, Ramoz cation between somatostatinergic axons and growth hormone-releasing
N, Grouselle D, Epelbaum J, Tolle V. Ghrelin-derived peptides: A factor (GRF) synthesizing neurons in the arcuate nucleus of the rat.
link between appetite/reward, gh axis, and psychiatric disorders? Front Histochemistry 89: 247-252, 1988.
Endocrinol (Lausanne) 5: 163, 2014. 305. Liposits Z, Merchenthaler I, Reid JJ, Negro-Vilar A. Galanin-
283. Lal S, Martin JB, De la Vega CE, Friesen HG. Comparison of the immunoreactive axons innervate somatostatin-synthesizing neurons in
effect of apomorphine and L-DOPA on serum growth hormone levels the anterior periventricular nucleus of the rat. Endocrinology 132: 917-
in normal men. Clin Endocrinol (Oxf) 4: 277-285, 1975. 923, 1993.
284. Lal S, Tolis G, Martin SB, Brown GM, Guyda H. Effect of clonidine on 306. Lissitzky JC, Giraud P, Conte-Devolx B, Gillioz P, Boudouresque F,
growth hormone, prolactin, luteinizing hormone, follicle-stimulating Eskay RL, Oliver C. beta-Endorphin is present in high concentration
hormone, and thyroid-stimulating hormone in the serum of normal in the hypophysial portal vessels of rats. Neurosci Lett 19: 191-195,
men. J Clin Endocrinol Metab 41: 827-832, 1975. 1980.
285. Lamberts SW, Macleod RM. Regulation of prolactin secretion at the 307. Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying
level of the lactotroph. Physiol Rev 70: 279-318, 1990. null mutations of the genes encoding insulin-like growth factor I (Igf-1)
286. Lange KH, Larsson B, Flyvbjerg A, Dall R, Bennekou M, Rasmussen and type 1 IGF receptor (Igf1r). Cell 75: 59-72, 1993.
MH, Orskov H, Kjaer M. Acute growth hormone administration causes 308. Locatelli V, Torsello A, Redaelli M, Ghigo E, Massare F, Muller EE.
exaggerated increases in plasma lactate and glycerol during moderate Cholinergic agonist and antagonist drugs modulate the growth hormone
to high intensity bicycling in trained young men. J Clin Endocrinol response to growth hormone-releasing hormone in the rat: Evidence for
Metab 87: 4966-4975, 2002. mediation by somatostatin. J Endocrinol 111: 271-278, 1986.
287. Lanneau C, Bluet-Pajot MT, Zizzari P, Csaba Z, Dournaud P, Helboe 309. Lonberg U, Damm P, Andersson AM, Main KM, Chellakooty M,
L, Hoyer D, Pellegrini E, Tannenbaum GS, Epelbaum J, Gardette R. Lauenborg J, Skakkebaek NE, Juul A. Increase in maternal placental
Involvement of the Sst1 somatostatin receptor subtype in the intrahy- growth hormone during pregnancy and disappearance during parturi-
pothalamic neuronal network regulating growth hormone secretion: tion in normal and growth hormone-deficient pregnancies. Am J Obstet
An in vitro and in vivo antisense study. Endocrinology 141: 967-979, Gynecol 188: 247-251, 2003.
2000. 310. Lough DS, Muller LD, Kensinger RS, Griel LC, Jr., Azzara CD. Effect
288. LaPaglia N, Steiner J, Kirsteins L, Emanuele M, Emanuele N. Leptin of exogenous bovine somatotropin on mammary lipid metabolism and
alters the response of the growth hormone releasing factor- growth milk yield in lactating dairy cows. J Dairy Sci 72: 1469-1476, 1989.
hormone–insulin-like growth factor-I axis to fasting. J Endocrinol 159: 311. Lumpkin MD, Samson WK, McCann SM. Effects of intraventricular
79-83, 1998. growth hormone-releasing factor on growth hormone release: Further
289. Lapp CA, Tyler JM, Lee YS, Stachura ME. Autocrine-paracrine inhi- evidence for ultrashort loop feedback. Endocrinology 116: 2070-2074,
bition of growth hormone and prolactin production by GH3 cell- 1985.
conditioned medium. In Vitro Cell Dev Biol 25: 528-534, 1989. 312. Luque RM, Cordoba-Chacon J, Ibanez-Costa A, Gesmundo I, Grande
290. Laron Z. Laron syndrome (primary growth hormone resistance or insen- C, Gracia-Navarro F, Tena-Sempere M, Ghigo E, Gahete MD, Granata
sitivity): The personal experience 1958-2003. J Clin Endocrinol Metab R, Kineman RD, Castano JP. Obestatin plays an opposite role in
89: 1031-1044, 2004. the regulation of pituitary somatotrope and corticotrope function in
291. Laron Z. The GH-IGF1 axis and longevity. The paradigm of IGF1 female primates and male/female mice. Endocrinology 155: 1407-1417,
deficiency. Hormones (Athens) 7: 24-27, 2008. 2014.
292. Lauwers E, Landuyt B, Arckens L, Schoofs L, Luyten W. Obestatin 313. Luque RM, Huang ZH, Shah B, Mazzone T, Kineman RD. Effects
does not activate orphan G protein-coupled receptor GPR39. Biochem of leptin replacement on hypothalamic-pituitary growth hormone axis
Biophys Res Commun 351: 21-25, 2006. function and circulating ghrelin levels in ob/ob mice. Am J Physiol
293. Le Tissier PR, Carmignac DF, Lilley S, Sesay AK, Phelps CJ, Hous- Endocrinol Metab 292: E891-E899, 2007.
ton P, Mathers K, Magoulas C, Ogden D, Robinson IC. Hypothalamic 314. Luque RM, Kineman RD. Impact of obesity on the growth hormone
growth hormone-releasing hormone (GHRH) deficiency: Targeted abla- axis: Evidence for a direct inhibitory effect of hyperinsulinemia on
tion of GHRH neurons in mice using a viral ion channel transgene. Mol pituitary function. Endocrinology 147: 2754-2763, 2006.
Endocrinol 19: 1251-1262, 2005. 315. Machlin LJ, Horino M, Hertelendy F, Kipnis DM. Plasma growth hor-
294. Le Tissier PR, Hodson DJ, Lafont C, Fontanaud P, Schaeffer M, Mollard mone and insulin levels in the pig. Endocrinology 82: 369-376, 1968.
P. Anterior pituitary cell networks. Front Neuroendocrinol 33: 252-266, 316. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei
2012. H, Kim S, Lallone R, Ranganathan S, Kern PA, Friedman JM. Leptin
295. Lee DK, Nguyen T, O’Neill GP, Cheng R, Liu Y, Howard AD, levels in human and rodent-measurement of plasma eptin and ob RNA
Coulombe N, Tan CP, Tang-Nguyen AT, George SR, O’Dowd BF. in obese and weight reduced subjects. Nat Med 1: 1155-1161, 1995.
Discovery of a receptor related to the galanin receptors. FEBS Lett 446: 317. Maheshwari HG, Silverman BL, Dupuis J, Baumann G. Phenotype and
103-107, 1999. genetic analysis of a syndrome caused by an inactivating mutation in
296. Leidy JW, Jr., McDermott MT, Robbins RJ. Effect of hypophysec- the growth hormone-releasing hormone receptor: Dwarfism of Sindh.
tomy and growth hormone administration on hypothalamic growth J Clin Endocrinol Metab 83: 4065-4074, 1998.
hormone-releasing hormone and somatostatin content: Relationship to 318. Maiter DM, Gabriel SM, Koenig JI, Russell WE, Martin JB. Sex-
age-related growth rate. Neuroendocrinology 51: 400-405, 1990. ual differentiation of growth hormone feedback effects on hypothala-
297. Li C, Chen P, Smith MS. Corticotropin releasing hormone neurons in mic growth hormone-releasing hormone and somatostatin. Neuroen-
the paraventricular nucleus are direct targets for neuropeptide Y neurons docrinology 51: 174-180, 1990.
in the arcuate nucleus: An anterograde tracing study. Brain Res 854: 319. Maiter DM, Hooi SC, Koenig JI, Martin JB. Galanin is a physiological
122-129, 2000. regulator of spontaneous pulsatile secretion of growth hormone in the
298. Li Q, Ren J. Influence of cardiac-specific overexpression of insulin-like male rat. Endocrinology 126: 1216-1222, 1990.
growth factor 1 on lifespan and aging-associated changes in cardiac 320. Mani BK, Walker AK, Lopez Soto EJ, Raingo J, Lee CE, Perello
intracellular Ca2 +homeostasis, protein damage and apoptotic protein M, Andrews ZB, Zigman JM. Neuroanatomical characterization of
expression. Aging cell 6: 799-806, 2007. a growth hormone secretagogue receptor-green fluorescent protein
299. Lima L, Arce V, Tresguerres JA, Devesa J. Clonidine potentiates the reporter mouse. J Comp Neurol 522: 3644-3666, 2014.
growth hormone (GH) response to GH-releasing hormone in nore- 321. Mansour A, Fox CA, Burke S, Meng F, Thompson RC, Akil H, Watson
pinephrine synthesis-inhibited rats: Evidence for an alpha-2-adrenergic SJ. Mu, delta, and kappa opioid receptor mRNA expression in the rat
control of hypothalamic release of somatostatin. Neuroendocrinology CNS: An in situ hybridization study. J Comp Neurol 350: 412-438,
57: 1155-1160, 1993. 1994.

728 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

322. Mansour A, Khachaturian H, Lewis ME, Akil H, Watson SJ. Autora- growth hormone-releasing factor (GRF). Endocrinology 114: 1950-
diographic differentiation of mu, delta, and kappa opioid receptors in 1952, 1984.
the rat forebrain and midbrain. J Neurosci 7: 2445-2464, 1987. 345. Miller KK. Endocrine dysregulation in anorexia nervosa update. J Clin
323. Marshall L, Molle M, Boschen G, Steiger A, Fehm HL, Born J. Greater Endocrinol Metab 96: 2939-2949, 2011.
efficacy of episodic than continuous growth hormone-releasing hor- 346. Milsom SR, Breier BH, Gallaher BW, Cox VA, Gunn AJ, Gluckman
mone (GHRH) administration in promoting slow-wave sleep (SWS). PD. Growth hormone stimulates galactopoiesis in healthy lactating
J Clin Endocrinol Metab 81: 1009-1013, 1996. women. Acta endocrinologica 127: 337-343, 1992.
324. Martha PM, Jr., Gorman KM, Blizzard RM, Rogol AD, Veldhuis JD. 347. Mims RB, Scott CL, Modebe OM, Bethune JE. Prevention of L-
Endogenous growth hormone secretion and clearance rates in normal dopa-induced growth hormone stimulation by hyperglycemia. J Clin
boys, as determined by deconvolution analysis: Relationship to age, Endocrinol Metab 37: 660-663, 1973.
pubertal status, and body mass. J Clin Endocrinol Metab 74: 336-344, 348. Minami S, Kamegai J, Hasegawa O, Sugihara H, Okada K, Wak-
1992. abayashi I. Expression of growth hormone receptor gene in rat hypotha-
325. Martha PM, Jr., Rogol AD, Veldhuis JD, Blizzard RM. A longitudinal lamus. J Neuroendocrinol 5: 691-696, 1993.
assessment of hormonal and physical alterations during normal puberty 349. Minami S, Kamegai J, Sugihara H, Suzuki N, Wakabayashi I. Growth
in boys. III. The neuroendocrine growth hormone axis during late pre- hormone inhibits its own secretion by acting on the hypothalamus
puberty. J Clin Endocrinol Metab 81: 4068-4074, 1996. through its receptors on neuropeptide Y neurons in the arcuate nucleus
326. Martin JB, Brazeau P, Tannenbaum GS, Willoughby JO, Epelbaum and somatostatin neurons in the periventricular nucleus. Endocr J
J, Terry LC, Durand D. Neuroendocrine organization of growth hor- 45(Suppl): S19-26, 1998.
mone regulation. Res Publ Assoc Res Nerv Ment Dis 56: 329-357, 350. Mirlesse V, Frankenne F, Alsat E, Poncelet M, Hennen G, Evain-
1978. Brion D. Placental growth hormone levels in normal pregnancy and
327. Martin JB, Durand D, Gurd W, Faille G, Audet J, Brazeau P. Neu- in pregnancies with intrauterine growth retardation. Pediatr Res 34:
ropharmacological regulation of episodic growth hormone and prolactin 439-442, 1993.
secretion in the rat. Endocrinology 102: 106-113, 1978. 351. Mitsugi N, Arita J, Kimura F. Effects of intracerebroventricular
328. Martinelli CE, Keogh JM, Greenfield JR, Henning E, van der Klaauw administration of growth hormone-releasing factor and corticotropin-
AA, Blackwood A, O’Rahilly S, Roelfsema F, Camacho-Hubner C, Pijl releasing factor on somatostatin secretion into rat hypophysial portal
H, Farooqi IS. Obesity due to melanocortin 4 receptor (MC4R) defi- blood. Neuroendocrinology 51: 93-96, 1990.
ciency is associated with increased linear growth and final height, fast- 352. Molero-Conejo E, Morales LM, Fernandez V, Raleigh X, Casanova A,
ing hyperinsulinemia, and incompletely suppressed growth hormone Connell L, Gomnez ME, Ryder E, Campos G. Serum insulin, leptin
secretion. J Clin Endocrinol Metab 96: E181-188, 2011. and growth hormone levels are associated with body mass index and
329. Mathias S, Held K, Ising M, Weikel JC, Yassouridis A, Steiger A. obesity index in adolescents. Arch Latinoam Nutr 56: 29-35, 2006.
Systemic growth hormone-releasing hormone (GHRH) impairs sleep 353. Molitch ME, Hlivyak LE. Growth hormone short-loop feedback:
in healthy young women. Psychoneuroendocrinology 32: 1021-1027, Anatomic specificity of growth hormone stimulation of hypothalamic
2007. somatostatin concentration. Horm Metab Res 12: 559-560, 1980.
330. Mauras N, Rogol AD, Veldhuis JD. Specific, time-dependent actions 354. Moller N, Jorgensen JO, Abildgard N, Orskov L, Schmitz O, Chris-
of low-dose ethinyl estradiol administration on the episodic release tiansen JS. Effects of growth hormone on glucose metabolism. Horm
of growth hormone, follicle-stimulating hormone, and luteinizing hor- Res 36(Suppl 1): 32-35, 1991.
mone in prepubertal girls with Turner’s syndrome. J Clin Endocrinol 355. Moreno-Reyes R, Kerkhofs M, L’Hermite-Baleriaux M, Thorner MO,
Metab 69: 1053-1058, 1989. Van Cauter E, Copinschi G. Evidence against a role for the growth
331. McIntyre HB, Odell WD. Physiological control of growth hormone in hormone-releasing peptide axis in human slow-wave sleep regulation.
the rabbit. Neuroendocrinology 16: 8-21, 1974. Am J Physiol 274: E779-784, 1998.
332. McKee KK, Tan CP, Palyha OC, Liu J, Feighner SD, Hreniuk DL, Smith 356. Mori K, Kim J, Sasaki K. Electrophysiological effect of ghrelin and
RG, Howard AD, Van der Ploeg LH. Cloning and characterization of somatostatin on rat hypothalamic arcuate neurons in vitro. Peptides 31:
two human G protein-coupled receptor genes (GPR38 and GPR39) 1139-1145, 2010.
related to the growth hormone secretagogue and neurotensin receptors. 357. Morita S, Yamashita S, Melmed S. Insulin-like growth factor I action on
Genomics 46: 426-434, 1997. rat anterior pituitary cells: Effects of intracellular messengers on growth
333. Melander T, Kohler C, Nilsson S, Hokfelt T, Brodin E, Theodorsson hormone secretion and messenger ribonucleic acid levels. Endocrinol-
E, Bartfai T. Autoradiographic quantitation and anatomical mapping ogy 121: 2000-2006, 1987.
of 125I-galanin binding sites in the rat central nervous system. J Chem 358. Moriwaki A, Wang JB, Svingos A, van Bockstaele E, Cheng P, Pickel
Neuroanat 1: 213-233, 1988. V, Uhl GR. mu Opiate receptor immunoreactivity in rat central nervous
334. Melmed S. Insulin suppresses growth hormone secretion by rat pituitary system. Neurochem Res 21: 1315-1331, 1996.
cells. J Clin Invest 73: 1425-1433, 1984. 359. Morselli LL, Nedeltcheva A, Leproult R, Spiegel K, Martino E, Legros
335. Mendelson WB, Slater S, Gold P, Gillin JC. The effect of growth JJ, Weiss RE, Mockel J, Van Cauter E, Copinschi G. Impact of GH
hormone administration on human sleep: A dose-response study. Biol replacement therapy on sleep in adult patients with GH deficiency of
Psychiatry 15: 613-618, 1980. pituitary origin. Eur J Endocrinol 168: 763-770, 2013.
336. Merchenthaler I. The hypophysiotropic galanin system of the rat brain. 360. Mounier F, Pellegrini E, Kordon C, Epelbaum J, Bluet-Pajot MT. Con-
Neuroscience 44: 643-654, 1991. tinuous intracerebroventricular administration of a corticotropin releas-
337. Merchenthaler I, Arimura A. Effect of hypophysectomy on immunocy- ing hormone antagonist amplifies spontaneous growth hormone pulses
tochemically demonstrated growth hormone releasing factor (GHRF) in the rat. J Endocrinol 152: 431-436, 1997.
in the rat brain. Peptides 6: 865-867, 1985. 361. Muccioli G, Papotti M, Locatelli V, Ghigo E, Deghenghi R. Binding
338. Merchenthaler I, Lopez FJ, Negro-Vilar A. Anatomy and physiology of 125I-labeled ghrelin to membranes from human hypothalamus and
of central galanin-containing pathways. Prog Neurobiol 40: 711-769, pituitary gland. J Endocrinol Invest 24: RC7-RC9, 2001.
1993. 362. Muller EE. Neural control of somatotropic function. Physiol Rev 67:
339. Meyer RM, Burgos-Robles A, Liu E, Correia SS, Goosens KA. A 962-1053, 1987.
ghrelin-growth hormone axis drives stress-induced vulnerability to 363. Muller EE, Cella SG, Parenti M, Deghenghi R, Locatelli V, De Gennaro
enhanced fear. Mol Psychiatry 19: 1284-1294, 2014. Colonna V, Torsello A, Cocchi D. Somatotropic dysregulation in old
340. Michener SR, Aimone LD, Yaksh TL, Go VL. Distribution of galanin- mammals. Horm Res 43: 39-45, 1995.
like immunoreactivity in the pig, rat and human central nervous system. 364. Mullis PE, Tonella P. Regulation of fetal growth: Consequences and
Peptides 11: 1217-1223, 1990. impact of being born small. Best Pract Res Clin Endocrinol Metab 22:
341. Miell JP, Corder R, Pralong FP, Gaillard RC. Effects of dexamethasone 173-190, 2008.
on growth hormone (GH)-releasing hormone, arginine- and dopamin- 365. Murakami Y, Kato Y, Kabayama Y, Tojo K, Inoue T, Imura H. Involve-
ergic stimulated GH secretion, and total plasma insulin-like growth ment of growth hormone (GH)-releasing factor in GH secretion induced
factor-I concentrations in normal male volunteers. J Clin Endocrinol by serotoninergic mechanisms in conscious rats. Endocrinology 119:
Metab 72: 675-681, 1991. 1089-1092, 1986.
342. Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP, 366. Murakami Y, Kato Y, Koshiyama H, Inoue T, Yanaihara N, Imura H.
Lanfrancone L, Pelicci PG. The p66shc adaptor protein controls oxida- Galanin stimulates growth hormone (GH) secretion via GH-releasing
tive stress response and life span in mammals. Nature 402: 309-313, factor (GRF) in conscious rats. Eur J Pharmacol 136: 415-418,
1999. 1987.
343. Miki N, Ono M, Miyoshi H, Tsushima T, Shizume K. Hypothalamic 367. Murakami Y, Kato Y, Shimatsu A, Koshiyama H, Hattori N, Yanai-
growth hormone-releasing factor (GRF) participates in the negative hara N, Imura H. Possible mechanisms involved in growth hormone
feedback regulation of growth hormone secretion. Life Sci 44: 469- secretion induced by galanin in the rat. Endocrinology 124: 1224-1229,
476, 1989. 1989.
344. Miki N, Ono M, Shizume K. Evidence that opiatergic and alpha- 368. Murphy LJ, Tachibana K, Friesen HG. Stimulation of hepatic insulin-
adrenergic mechanisms stimulate rat growth hormone release via like growth factor-I gene expression by ovine prolactin: Evidence for

Volume 6, April 2016 729


Growth Hormone Axis Comprehensive Physiology

intrinsic somatogenic activity in the rat. Endocrinology 122: 2027-2033, 392. Oosterom R, Verleun T, Zuiderwijk J, Lamberts SW. Growth hormone
1988. secretion by cultured rat anterior pituitary cells. Effects of culture con-
369. Myers MG, Cowley MA, Munzberg H. Mechanisms of leptin action ditions and dexamethasone. Endocrinology 113: 735-741, 1983.
and leptin resistance. Annu Rev Physiol 70: 537-556, 2008. 393. Osterstock G, Escobar P, Mitutsova V, Gouty-Colomer L-A, Fontanaud
370. Nakagawa K, Akikawa K, Matsubara M, Kubo M. Effect of dexametha- P, Molino F, Fehrentz J-A, Carmignac D, Martinez J, Guerineau NC,
sone on growth hormone (GH) response to growth hormone releasing Robinson ICAF, Mollard P, Méry P-F. Ghrelin stimulation of growth
hormone in acromegaly. J Clin Endocrinol Metab 60: 306-310, 1985. hormone-releasing hormone neurons is direct in the arcuate nucleus.
371. Nakagawa K, Akikawa K, Matsubara M, Kubo M, Ishizuka T, Obara T. PLoS ONE 5: e9159, 2010.
Biphasic effects of dexamethasone on growth hormone release in vitro. 394. Ottlecz A, Snyder GD, McCann SM. Regulatory role of galanin in
Endocrinol Jpn 34: 947-953, 1987. control of hypothalamic-anterior pituitary function. Proc Natl Acad Sci
372. Nakamura S, Mizuno M, Katakami H, Gore AC, Terasawa E. Aging- U S A 85: 9861-9865, 1988.
related changes in in vivo release of growth hormone-releasing hormone 395. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill
and somatostatin from the stalk-median eminence in female rhesus GF, Jr. Brain metabolism during fasting. J Clin Invest 46: 1589-1595,
monkeys (Macaca mulatta). J Clin Endocrinol Metab 88: 827-833, 1967.
2003. 396. Ozata M, Ozdemir IC, Licinio J. Human leptin deficiency caused by
373. Nakazato M, Murakami N, Date Y, Kojima M, Matsuo H, Kangawa a missense mutation: Multiple endocrine defects, decreased sympa-
K, Matsukura S. A role for ghrelin in the central regulation of feeding. thetic tone, and immune system dysfunction indicate new targets for
Nature 409: 194-198, 2001. leptin action, greater central than peripheral resistance to the effects of
374. Nam SY, Lee EJ, Kim KR, Lee HC, Nam MS, Cho JH, Huh KB. Long- leptin, and spontaneous correction of leptin-mediated defects. J Clin
term administration of acipimox potentiates growth hormone response Endocrinol Metab 84: 3686-3695, 1999.
to growth hormone-releasing hormone by decreasing serum free fatty 397. Palkovits M, Rokaeus A, Antoni FA, Kiss A. Galanin in the
acid in obesity. Metabolism 45: 594-597, 1996. hypothalamo-hypophyseal system. Neuroendocrinology 46: 417-423,
375. Nam SY, Marcus C. Growth hormone and adipocyte function in obesity. 1987.
Horm Res 53(Suppl 1): 87-97, 2000. 398. Palmiter RD, Norstedt G, Gelinas RE, Hammer RE, Brinster RL.
376. Nass R, Farhy LS, Liu J, Pezzoli SS, Johnson ML, Gaylinn BD, Thorner Metallothionein-human GH fusion genes stimulate growth of mice.
MO. Age-dependent decline in acyl-ghrelin concentrations and reduced Science 222: 809-814, 1983.
association of acyl-ghrelin and growth hormone in healthy older adults. 399. Pan W, Yu Y, Cain CM, Nyberg F, Couraud PO, Kastin AJ. Permeation
J Clin Endocrinol Metab 99: 602-608, 2014. of growth hormone across the blood-brain barrier. Endocrinology 146:
377. Nass R, Farhy LS, Liu J, Prudom CE, Johnson ML, Veldhuis P, Pezzoli 4898-4904, 2005.
SS, Oliveri MC, Gaylinn BD, Geysen HM, Thorner MO. Evidence for 400. Pantel J, Legendre M, Cabrol S, Hilal L, Hajaji Y, Morisset S, Nivot
acyl-ghrelin modulation of growth hormone release in the fed state. S, Vie-Luton M-P, Grouselle D, de Kerdanet M, Kadiri A, Epelbaum
J Clin Endocrinol Metab 93: 1988-1994, 2008. J, Le Bouc Y, Amselem S. Loss of constitutive activity of the growth
378. Ndon JA, Giustina A, Wehrenberg WB. Hypothalamic regulation of hormone secretagogue receptor in familial short stature. J Clin Invest
impaired growth hormone secretion in diabetic rats. 1. Studies in 116: 760-768, 2006.
streptozotocin-induced diabetic rats. Neuroendocrinology 55: 500-505, 401. Pantel J, Legendre M, Nivot S, Morisset S, Vie-Luton M-P, Le Bouc Y,
1992. Epelbaum J, Amselem S. Recessive isolated growth hormone deficiency
379. Neggers SJ, van der Lely AJ. Somatostatin analog and pegvisomant and mutations in the ghrelin receptor. J Clin Endocrinol Metab 94:
combination therapy for acromegaly. Nat Rev Endocrinol 5: 546-552, 4334-4341, 2009.
2009. 402. Papadakis MA, Grady D, Black D, Tierney MJ, Gooding GA, Scham-
380. Niimi M, Takahara J, Sato M, Kawanishi K. Immunohistochemi- belan M, Grunfeld C. Growth hormone replacement in healthy older
cal identification of galanin and growth hormone-releasing factor- men improves body composition but not functional ability. Ann Intern
containing neurons projecting to the median eminence of the rat. Neu- Med 124: 708-716, 1996.
roendocrinology 51: 572-575, 1990. 403. Parrella E, Longo VD. Insulin/IGF-I and related signaling pathways
381. Nogueiras R, Pfluger P, Tovar S, Arnold M, Mitchell S, Morris A, Perez- regulate aging in nondividing cells: From yeast to the mammalian brain.
Tilve D, Vázquez MJ, Wiedmer P, Castañeda TR, DiMarchi R, Tschöp ScientificWorldJournal 10: 161-177, 2010.
M, Schurmann A, Joost H-G, Williams LM, Langhans W, Diéguez C. 404. Patchett AA, Nargund RP, Tata JR, Chen MH, Barakat KJ, Johnston
Effects of obestatin on energy balance and growth hormone secretion DB, Cheng K, Chan WW, Butler B, Hickey G, Jacks T, Schleim K,
in rodents. Endocrinology 148: 21-26, 2007. Pong SS, Chaung LYP, Chen HY, Frazier E, Leung KH, Chiu SHL,
382. Nolte W, Radisch C, Rodenbeck A, Wiltfang J, Hufner M. Polysomno- Smith RG. Design and biological activities of L-163,191 (MK-0677):
graphic findings in five adult patients with pituitary insufficiency before A potent, orally active growth hormone secretagogue. Proc Natl Acad
and after cessation of human growth hormone replacement therapy. Clin Sci U S A 92: 7001-7005, 1995.
Endocrinol (Oxf) 56: 805-810, 2002. 405. Patel YC, Wheatley T, Zingg HH. Increased blood somatostatin con-
383. Obal F, Jr., Alt J, Taishi P, Gardi J, Krueger JM. Sleep in mice with non- centration in streptozotocin diabetic rats. Life Sci 27: 1563-1570, 1980.
functional growth hormone-releasing hormone receptors. Am J Physiol 406. Pazos Y, Casanueva FF, Camiña JP. Basic aspects of ghrelin action.
Regul Integr Comp Physiol 284: R131-R139, 2003. Vitam Horm 77: 89-119, 2008.
384. Obal F, Jr., Fang J, Taishi P, Kacsoh B, Gardi J, Krueger JM. Defi- 407. Pedragosa-Badia X, Stichel J, Beck-Sickinger AG. Neuropeptide Y
ciency of growth hormone-releasing hormone signaling is associated receptors: How to get subtype selectivity. Front Endocrinol (Lausanne)
with sleep alterations in the dwarf rat. J Neurosci 21: 2912-2918, 2001. 4: 5, 2013.
385. Obal F, Jr., Floyd R, Kapas L, Bodosi B, Krueger JM. Effects of sys- 408. Peker Y, Svensson J, Hedner J, Grote L, Johannsson G. Sleep apnoea
temic GHRH on sleep in intact and hypophysectomized rats. Am J and quality of life in growth hormone (GH)-deficient adults before and
Physiol 270: E230-237, 1996. after 6 months of GH replacement therapy. Clin Endocrinol (Oxf) 65:
386. Obal F, Jr., Krueger JM. GHRH and sleep. Sleep Med Rev 8: 367-377, 98-105, 2006.
2004. 409. Peroni CN, Hayashida CY, Nascimento N, Longuini VC, Toledo RA,
387. Ohyama T, Sato M, Niimi M, Hizuka N, Takahara J. Effects of short- Bartolini P, Bowers CY, Toledo SP. Growth hormone response to
and long-term dexamethasone treatment on growth and growth hor- growth hormone-releasing peptide-2 in growth hormone-deficient little
mone (GH)-releasing hormone (GRH)-GH-insulin-like growth factor-I mice. Clinics (Sao Paulo) 67: 265-272, 2012.
axis in conscious rats. Endocr J 44: 827-835, 1997. 410. Perras B, Marshall L, Kohler G, Born J, Fehm HL. Sleep and endocrine
388. Okimura Y, Ukai K, Hosoda H, Murata M, Iguchi G, Iida K, Kaji H, changes after intranasal administration of growth hormone-releasing
Kojima M, Kangawa K, Chihara K. The role of circulating ghrelin in hormone in young and aged humans. Psychoneuroendocrinology 24:
growth hormone (GH) secretion in freely moving male rats. Life Sci 743-757, 1999.
72: 2517-2524, 2003. 411. Peterfreund RA, Vale WW. Somatostatin analogs inhibit somatostatin
389. Olchovsky D, Bruno JF, Wood TL, Gelato MC, Leidy JW, Jr., Gilbert secretion from cultured hypothalamus cells. Neuroendocrinology 39:
JM, Jr., Berelowitz M. Altered pituitary growth hormone (GH) regula- 397-402, 1984.
tion in streptozotocin-diabetic rats: A combined defect of hypothalamic 412. Pfluger PT, Kirchner H, Günnel S, Schrott B, Perez-Tilve D, Fu S,
somatostatin and GH-releasing factor. Endocrinology 126: 53-61, 1990. Benoit SC, Horvath T, Joost H-G, Wortley KE, Sleeman MW, Tschöp
390. Ono M, Miki N, Murata Y, Osaki E, Tamitsu K, Ri T, Yamada MH. Simultaneous deletion of ghrelin and its receptor increases motor
M, Demura H. Sexually dimorphic expression of pituitary growth activity and energy expenditure. Am J Physiol Gastrointest Liver Phys-
hormone-releasing factor receptor in the rat. Biochem Biophys Res iol 294: G610-G618, 2008.
Commun 216: 1060-1066, 1995. 413. Pierroz DD, Catzeflis C, Aebi AC, Rivier JE, Aubert ML. Chronic
391. Ono N, Lumpkin MD, Samson WK, McDonald JK, McCann SM. administration of neuropeptide Y into the lateral ventricle inhibits both
Intrahypothalamic action of corticotrophin-releasing factor (CRF) to the pituitary-testicular axis and growth hormone and insulin-like growth
inhibit growth hormone and LH release in the rat. Life Sci 35: 1117- factor I secretion in intact adult male rats. Endocrinology 137: 3-12,
1123, 1984. 1996.

730 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

414. Pietschmann P, Schernthaner G, Prskavec F, Gisinger C, Freyler H. No 435. Riskind PN, Millard WJ, Martin JB. Opiate modulation of the anterior
evidence for increased growth hormone responses to growth hormone- pituitary hormone response during suckling in the rat. Endocrinology
releasing hormone in patients with diabetic retinopathy. Diabetes 36: 114: 1232-1237, 1984.
159-162, 1987. 436. Rivier C, Vale W. Involvement of corticotropin-releasing factor and
415. Plotsky PM, Vale W. Patterns of growth hormone-releasing factor and somatostatin in stress-induced inhibition of growth hormone secretion
somatostatin secretion into the hypophysial-portal circulation of the rat. in the rat. Endocrinology 117: 2478-2482, 1985.
Science 230: 461-463, 1985. 437. Rivier C, Vale W, Ling N, Brown M, Guillemin R. Stimulation in vivo
416. Polkowska J, Wankowska M, Romanowicz K, Gajewska A, Misztal T, of the secretion of prolactin and growth hormone by beta-endorphin.
Wojcik-Gladysz A. The effect of intracerebroventricular infusions of Endocrinology 100: 238-241, 1977.
ghrelin and/or short fasting on the gene expression and immunoreactiv- 438. Robinson IC, Hindmarsh PC. The growth hormone secretory pattern
ity of somatostatin in the hypothalamic neurons and on pituitary growth and statural growth. In: Kostyo JL, editor. Handbook of Physiology.
hormone in prepubertal female lambs. Morphological arguments. Brain New York, Oxford: Oxford University Press for the American Physiol-
Res 1414: 41-49, 2011. ogy Society, 1999, pp. 329-396.
417. Pontiroli AE, Lanzi R, Monti LD, Sandoli E, Pozza G. Growth hormone 439. Roh SG, Clarke IJ, Xu RW, Goding JW, Loneragan K, Chen C.
(GH) autofeedback on GH response to GH-releasing hormone. Role of The in vitro effect of leptin on basal and growth hormone-releasing
free fatty acids and somatostatin. J Clin Endocrinol Metab 72: 492-495, hormone-stimulatedgrowth hormone secretion from the ovine pituitary
1991. gland. Neuroendocrinology 68: 361-364, 1998.
418. Pontiroli AE, Manzoni MF, Malighetti ME, Lanzi R. Restoration of 440. Roh S-G, Nie G-Y, Loneragan K, Gertler A, Chen C. Direct modifica-
growth hormone (GH) response to GH-releasing hormone in elderly tion of somatotrope function by long-term leptin treatment of primary
and obese subjects by acute pharmacological reduction of plasma free cultured ovine pituitary cells. Endocrinology 142: 5167-5171, 2001.
fatty acids. J Clin Endocrinol Metab 81: 3998-4001, 1996. 441. Rose SR, Municchi G, Barnes KM, Kamp GA, Uriarte MM, Ross
419. Pralong FP, Miell JP, Corder R, Gaillard RC. Dexamethasone treatment JL, Cassorla F, Cutler GB, Jr. Spontaneous growth hormone secretion
in man induces changes in 24-hour growth hormone (GH) secretion increases during puberty in normal girls and boys. J Clin Endocrinol
profile without altering total GH released. J Clin Endocrinol Metab 73: Metab 73: 428-435, 1991.
1191-1196, 1991. 442. Rose SR, Ross JL, Uriarte M, Barnes KM, Cassorla FG, Cutler GB, Jr.
420. Quabbe HJ, Schilling E, Helge H. Pattern of growth hormone secretion The advantage of measuring stimulated as compared with spontaneous
during a 24-hour fast in normal adults. J Clin Endocrinol Metab 26: growth hormone levels in the diagnosis of growth hormone deficiency.
1173-1177, 1966. N Engl J Med 319: 201-207, 1988.
421. Quaife CJ, Mathews LS, Pinkert CA, Hammer RE, Brinster RL, 443. Rosenbloom AL, Guevara-Aguirre J. Lessons from the genetics of laron
Palmiter RD. Histopathology associated with elevated levels of syndrome. Trends Endocrinol Metab 9: 276-283, 1998.
growth hormone and insulin-like growth factor I in transgenic mice. 444. Rosenthal SM, Silverman BL, Wehrenberg WB. Exogenous growth hor-
Endocrinology 124: 40-48, 1989. mone inhibits bovine but not murine pituitary growth hormone secre-
422. Gardette R, Petit F, Peineau S, Lanneau C, Epelbaum J. Coxpression tion in vitro: Evidence for a direct feedback of growth hormone on the
of multiple somatostatin receptors in individual cells. In: Srikant C, pituitary. Neuroendocrinology 53: 597-600, 1991.
Somatostatin. The Netherlands: Kluwer Academics Publishers, 2004, 445. Roth J, Glick SM, Yalow RS, Bersonsa. Hypoglycemia: A potent stim-
pp. 123-142. ulus to secretion of growth hormone. Science 140: 987-988, 1963.
423. Radomski MW, Buguet A, Montmayeur A, Bogui P, Bourdon L, Doua 446. Ruch W, Jaton AL, Bucher B, Marbach P, Doepfner W. Alpha adren-
F, Lonsdorfer A, Tapie P, Dumas M. Twenty-four-hour plasma cortisol ergic control of growth hormone in adult male rats. Experientia 32:
and prolactin in human African trypanosomiasis patients and healthy 529-531, 1976.
African controls. Am J Trop Med Hyg 52: 281-286, 1995. 447. Rudman D, Feller AG, Nagraj HS, Gergans GA, Lalitha PY, Goldberg
424. Raposinho PD, Castillo E, d’Alleves V, Broqua P, Pralong FP, Aubert AF, Schlenker RA, Cohn L, Rudman IW, Mattson DE. Effects of human
ML. Chronic blockade of the melanocortin 4 receptor subtype leads to growth hormone in men over 60 years old. N Engl J Med 323: 1-6, 1990.
obesity independently of neuropeptide Y action, with no adverse effects 448. Sainsbury A, Schwarzer C, Couzens M, Fetissov S, Furtinger S, Jenkins
on the gonadotropic and somatotropic axes. Endocrinology 141: 4419- A, Cox HM, Sperk G, Hokfelt T, Herzog H. Important role of hypotha-
4427, 2000. lamic Y2 receptors in body weight regulation revealed in conditional
425. Raposinho PD, Pierroz DD, Broqua P, White RB, Pedrazzini T, Aubert knockout mice. Proc Natl Acad Sci U S A 99: 8938-8943, 2002.
ML. Chronic administration of neuropeptide Y into the lateral ventri- 449. Sakharova AA, Horowitz JF, Surya S, Goldenberg N, Harber MP,
cle of C57BL/6J male mice produces an obesity syndrome including Symons K, Barkan A. Role of growth hormone in regulating lipoly-
hyperphagia, hyperleptinemia, insulin resistance, and hypogonadism. sis, proteolysis, and hepatic glucose production during fasting. J Clin
Mol Cell Endocrinol 185: 195-204, 2001. Endocrinol Metab 93: 2755-2759, 2008.
426. Rasmussen MH, Hvidberg A, Juul A, Main KM, Gotfredsen A, Skakke- 450. Saleri R, Giustina A, Tamanini C, Valle D, Burattin A, Wehrenberg
baek NE, Hilsted J, Skakkebae NE. Massive weight loss restores 24- WB, Baratta M. Leptin stimulates growth hormone secretion via a
hour growth hormone release profiles and serum insulin-like growth direct pituitary effect combined with a decreased somatostatin tone in
factor-I levels in obese subjects. J Clin Endocrinol Metab 80: 1407- a median eminence-pituitary perifusion study. Neuroendocrinology 79:
1415, 1995. 221-228, 2004.
427. Redwine L, Hauger RL, Gillin JC, Irwin M. Effects of sleep and 451. Salvatori R, Hayashida CY, Aguiar-Oliveira MH, Phillips JA, III, Souza
sleep deprivation on interleukin-6, growth hormone, cortisol, and mela- AH, Gondo RG, Toledo SP, Conceicao MM, Prince M, Maheshwari
tonin levels in humans. J Clin Endocrinol Metab 85: 3597-3603, HG, Baumann G, Levine MA. Familial dwarfism due to a novel muta-
2000. tion of the growth hormone-releasing hormone receptor gene. J Clin
428. Reichenbach A, Steyn FJ, Sleeman MW, Andrews ZB. Ghrelin recep- Endocrinol Metab 84: 917-923, 1999.
tor expression and colocalization with anterior pituitary hormones 452. Sanchez-Cardenas C, Fontanaud P, He Z, Lafont C, Meunier AC, Scha-
using a GHSR-GFP mouse line. Endocrinology 153: 5452-5466, effer M, Carmignac D, Molino F, Coutry N, Bonnefont X, Gouty-
2012. Colomer LA, Gavois E, Hodson DJ, Le Tissier P, Robinson IC, Mollard
429. Rettori V, Milenkovic L, Aguila MC, McCann SM. Physiologically P. Pituitary growth hormone network responses are sexually dimorphic
significant effect of neuropeptide Y to suppress growth hormone release and regulated by gonadal steroids in adulthood. Proc Natl Acad Sci U
by stimulating somatostatin discharge. Endocrinology 126: 2296-2301, S A 107: 21878-21883, 2010.
1990. 453. Sato M, Frohman LA. Differential effects of central and peripheral
430. Richards NT, Wood SM, Christofides ND, Bhuttacharji SC, Bloom SR. administration of growth hormone (GH) and insulin-like growth factor
Impaired growth hormone response to human pancreatic growth hor- on hypothalamic GH-releasing hormone and somatostatin gene expres-
mone releasing factor [GRF(1-44)] in type 2 (non-insulin-dependent) sion in GH-deficient dwarf rats. Endocrinology 133: 793-799, 1993.
diabetes. Diabetologia 27: 529-534, 1984. 454. Sato M, Takahara J, Niimi M, Irino S. Effects of somatostatin on the
431. Richardson SB, Hollander CS, D’Eletto R, Greenleaf PW, Thaw C. growth hormone-releasing factor-induced growth hormone secretion
Acetylcholine inhibits the release of somatostatin from rat hypothala- in rats with electrical and chemical inhibitions of endogenous growth
mus in vitro. Endocrinology 107: 122-129, 1980. hormone-releasing factor and somatostatin. J Neuroendocrinol 2: 555-
432. Richardson SB, Hollander CS, Prasad JA, Hirooka Y. Somatostatin 561, 1990.
release from rat hypothalamus in vitro: Effects of melatonin and sero- 455. Sato M, Takahara J, Niimi M, Tagawa R, Irino S. Characterization of
tonin. Endocrinology 109: 602-606, 1981. the stimulatory effect of galanin on growth hormone release from the
433. Richardson SB, Twente S. Inhibition of hypothalamic somatostatin rat anterior pituitary. Life Sci 48: 1639-1644, 1991.
release by beta-adrenergic antagonists. Endocrinology 126: 1043-1046, 456. Saunders A, Terry LC, Audet J, Brazeau P, Martin JB. Dynamic studies
1990. of growth hormone and prolactin secretion in the female rat. Neuroen-
434. Rimoin DL, Merimee TJ, Mc Kusick VA. Growth-hormone deficiency docrinology 21: 193-203, 1976.
in man: An isolated, recessively inherited defect. Science 152: 1635- 457. Savendahl L. The effect of acute and chronic stress on growth. Sci
1637, 1966. Signal 5: pt9, 2012.

Volume 6, April 2016 731


Growth Hormone Axis Comprehensive Physiology

458. Sawchenko PE, Pfeiffer SW. Ultrastructural localization of neuropep- 481. Stefaneanu L, Kovacs K, Horvath E, Losinski NE, Mayerhofer A, Wag-
tide Y and galanin immunoreactivity in the paraventricular nucleus of ner TE, Bartke A. An immunocytochemical and ultrastructural study
the hypothalamus in the rat. Brain Res 474: 231-245, 1988. of adenohypophyses of mice transgenic for human growth hormone.
459. Schioth HB, Muceniece R, Wikberg JE. Characterization of the binding Endocrinology 126: 608-615, 1990.
of MSH-B, HB-228, GHRP-6 and 153N-6 to the human melanocortin 482. Stefaneanu L, Kovacs K, Lloyd RV, Scheithauer BW, Young WF, Jr.,
receptor subtypes. Neuropeptides 31: 565-571, 1997. Sano T, Jin L. Pituitary lactotrophs and somatotrophs in pregnancy:
460. Schneider HJ, Oertel H, Murck H, Pollmacher T, Stalla GK, Steiger A. A correlative in situ hybridization and immunocytochemical study.
Night sleep EEG and daytime sleep propensity in adult hypopituitary Virchows Arch B Cell Pathol Incl Mol Pathol 62: 291-296, 1992.
patients with growth hormone deficiency before and after six months of 483. Steiger A. Sleep and endocrinology. J Intern Med 254: 13-22, 2003.
growth hormone replacement. Psychoneuroendocrinology 30: 29-37, 484. Steiger A, Guldner J, Hemmeter U, Rothe B, Wiedemann K, Holsboer
2005. F. Effects of growth hormone-releasing hormone and somatostatin on
461. Schonbrunn A. Glucocorticoids down-regulate somatostatin receptors sleep EEG and nocturnal hormone secretion in male controls. Neuroen-
on pituitary cells in culture. Endocrinology 110: 1147-1154, 1982. docrinology 56: 566-573, 1992.
462. Schuessler P, Uhr M, Ising M, Schmid D, Weikel J, Steiger A. Nocturnal 485. Steyn FJ. Nutrient sensing overrides somatostatin and GHRH to control
ghrelin levels–relationship to sleep EEG, the levels of growth hormone, pulsatile GH release. J Neuroendocrinol 2015.
ACTH and cortisol–and gender differences. J Sleep Res 14: 329-336, 486. Steyn FJ, Huang L, Ngo ST, Leong JW, Tan HY, Xie TY, Parlow
2005. AF, Veldhuis JD, Waters MJ, Chen C. Development of a method
463. Schussler P, Yassouridis A, Uhr M, Kluge M, Weikel J, Holsboer for the determination of pulsatile growth hormone secretion in mice.
F, Steiger A. Growth hormone-releasing hormone and corticotropin- Endocrinology 152: 3165-3171, 2011.
releasing hormone enhance non-rapid-eye-movement sleep after sleep 487. Steyn FJ, Leong JW, Huang L, Tan HY, Xie TY, Nelson C, Waters
deprivation. Am J Physiol Endocrinol Metab 291: E549-E556, 2006. MJ, Veldhuis JD, Epelbaum J, Chen C. GH does not modulate the early
464. Seifert H, Perrin M, Rivier J, Vale W. Binding sites for growth hormone fasting-induced release of free fatty acids in mice. Endocrinology 153:
releasing factor on rat anterior pituitary cells. Nature 313: 487-489, 273-282, 2012.
1985. 488. Steyn FJ, Xie TY, Huang L, Ngo ST, Veldhuis JD, Waters MJ, Chen C.
465. Seifert H, Perrin M, Rivier J, Vale W. Growth hormone-releasing factor Increased adiposity and insulin correlates with the progressive suppres-
binding sites in rat anterior pituitary membrane homogenates: Modu- sion of pulsatile GH secretion during weight gain. J Endocrinol 218:
lation by glucocorticoids. Endocrinology 117: 424-426, 1985. 233-244, 2013.
466. Seim I, Herington AC, Chopin LK. New insights into the molecular 489. Strauch G, Girault D, Rifai M, Bricaire H. Alpha-MSH stimulation
complexity of the ghrelin gene locus. Cytokine Growth Factor Rev 20: of growth hormone release. J Clin Endocrinol Metab 37: 990-993,
297-304, 2009. 1973.
467. Selman C, Lingard S, Choudhury AI, Batterham RL, Claret M, 490. Strauch G, Luton JP, Bricaire H. Somatotropin insufficiency in Cush-
Clements M, Ramadani F, Okkenhaug K, Schuster E, Blanc E, Piper ing’s syndrome: Regression after treatment of the hypercorticism. Ann
MD, Al-Qassab H, Speakman JR, Carmignac D, Robinson IC, Thornton Endocrinol (Paris) 31: 985-992, 1970.
JM, Gems D, Partridge L, Withers DJ. Evidence for lifespan extension 491. Strickland AL, Underwood LE, Voina SJ, French FS, Van Wyk JJ.
and delayed age-related biomarkers in insulin receptor substrate 1 null Growth retardation in Cushing’s syndrome. Am J Dis Child 123: 207-
mice. FASEB J 22: 807-818, 2008. 213, 1972.
468. Selman C, Partridge L, Withers DJ. Replication of extended lifespan 492. Suh Y, Atzmon G, Cho MO, Hwang D, Liu B, Leahy DJ, Barzilai N,
phenotype in mice with deletion of insulin receptor substrate 1. PLoS Cohen P. Functionally significant insulin-like growth factor I receptor
One 6: e16144, 2011. mutations in centenarians. Proc Natl Acad Sci U S A 105: 3438-3442,
469. Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury 2008.
AI, Claret M, Al-Qassab H, Carmignac D, Ramadani F, Woods A, 493. Sun LY, Spong A, Swindell WR, Fang Y, Hill C, Huber JA, Boehm
Robinson IC, Schuster E, Batterham RL, Kozma SC, Thomas G, Carling JD, Westbrook R, Salvatori R, Bartke A. Growth hormone-releasing
D, Okkenhaug K, Thornton JM, Partridge L, Gems D, Withers DJ. hormone disruption extends lifespan and regulates response to caloric
Ribosomal protein S6 kinase 1 signaling regulates mammalian life restriction in mice. Elife 2: e01098, 2013.
span. Science 326: 140-144, 2009. 494. Sun Y, Ahmed S, Smith RG. Deletion of ghrelin impairs neither growth
470. Sharp PS, Foley K, Chahal P, Kohner EM. The effect of plasma glucose nor appetite. Mol Cell Biol 23: 7973-7981, 2003.
on the growth hormone response to human pancreatic growth hormone 495. Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth
releasing factor in normal subjects. Clin Endocrinol (Oxf) 20: 497-501, hormone release and appetite is mediated through the growth hormone
1984. secretagogue receptor. Proc Natl Acad Sci USA 101: 4679-4684, 2004.
471. Shaw AM, Irani BG, Moore MC, Haskell-Luevano C, Millard WJ. 496. Suzuki N, Okada K, Minami S, Wakabayashi I. Inhibitory effect of
Ghrelin-induced food intake and growth hormone secretion are altered neuropeptide Y on growth hormone secretion in rats is mediated by
in melanocortin 3 and 4 receptor knockout mice. Peptides 26: 1720- both Y1- and Y2-receptor subtypes and abolished after anterolateral
1727, 2005. deafferentation of the medial basal hypothalamus. Regul Pept 65: 145-
472. Sheppard MC, Kronheim S, Pimstone BL. Stimulation by growth hor- 151, 1996.
mone of somatostatin release from the rat hypothalamus in vitro. Clin 497. Svensson J, Sjogren K, Faldt J, Andersson N, Isaksson O, Jansson JO,
Endocrinol (Oxf) 9: 583-586, 1978. Ohlsson C. Liver-derived IGF-I regulates mean life span in mice. PLoS
473. Shibasaki T, Yamauchi N, Hotta M, Masuda A, Imaki T, Demura One 6: e22640, 2011.
H, Ling N, Shizume K. In vitro release of growth hormone-releasing 498. Swanson LW, Sawchenko PE, Rivier J, Vale WW. Organization of
factor from rat hypothalamus: Effect of insulin-like growth factor-1. ovine corticotropin-releasing factor immunoreactive cells and fibers in
Regul Pept 15: 47-53, 1986. the rat brain: An immunohistochemical study. Neuroendocrinology 36:
474. Smith RG, Griffin PR, Xu Y, Smith AG, Liu K, Calacay J, Feighner SD, 165-186, 1983.
Pong C, Leong D, Pomés A, Cheng K, Van der Ploeg LH, Howard AD, 499. Szentirmai E, Kapas L, Sun Y, Smith RG, Krueger JM. Spontaneous
Schaeffer J, Leonard RJ. Adenosine: A partial agonist of the growth sleep and homeostatic sleep regulation in ghrelin knockout mice. Am J
hormone secretagogue receptor. Biochem Biophys Res Commun 276: Physiol Regul Integr Comp Physiol 293: R510-R517, 2007.
1306-1313, 2000. 500. Taguchi A, Wartschow LM, White MF. Brain IRS2 signaling coordi-
475. Smythe GA, Lazarus L. Suppression of human growth hormone secre- nates life span and nutrient homeostasis. Science 317: 369-372, 2007.
tion by melatonin and cyproheptadine. J Clin Invest 54: 116-121, 1974. 501. Takahashi K, Daughaday WH, Kipnis DM. Regulation of immunoreac-
476. Somm E, Bonnet N, Martinez A, Marks PM, Cadd VA, Elliott M, tive growth hormone secretion in male rats. Endocrinology 88: 909-917,
Toulotte A, Ferrari SL, Rizzoli R, Huppi PS, Harper E, Melmed S, Jones 1971.
R, Aubert ML. A botulinum toxin-derived targeted secretion inhibitor 502. Takaya K, Ariyasu H, Kanamoto N, Iwakura H, Yoshimoto A, Harada
downregulates the GH/IGF1 axis. J Clin Invest 122: 3295-3306, 2012. M, Mori K, Komatsu Y, Usui T, Shimatsu A, Ogawa Y, Hosoda K,
477. Spiess J, Rivier J, Rivier C, Vale W. Primary structure of corticotropin- Akamizu T, Kojima M, Kangawa K, Nakao K. Ghrelin strongly stimu-
releasing factor from ovine hypothalamus. Proc Natl Acad Sci U S A lates growth hormone release in humans. J Clin Endocrinol Metab 85:
78: 6517-6521, 1981. 4908-4911, 2000.
478. Stanley S, Domingos AI, Kelly L, Garfield A, Damanpour S, Heisler L, 503. Tan HY, Huang L, Simmons D, Veldhuis JD, Steyn FJ, Chen C.
Friedman J. Profiling of glucose-sensing neurons reveals that GHRH Hypothalamic distribution of somatostatin mRNA expressing neurones
neurons are activated by hypoglycemia. Cell Metab 18: 596-607, 2013. relative to pubertal and adult changes in pulsatile growth hormone
479. Stark R, Ashley SE, Andrews ZB. AMPK and the neuroendocrine secretion in mice. J Neuroendocrinol 25: 910-919, 2013.
regulation of appetite and energy expenditure. Mol Cell Endocrinol 504. Tannenbaum GS. Growth hormone-releasing factor: Direct effects on
366: 215-223, 2013. growth hormone, glucose, and behavior via the brain. Science 226:
480. Stefaneanu L, Kovacs K, Bartke A, Mayerhofer A, Wagner TE. Pituitary 464-466, 1984.
morphology of transgenic mice expressing bovine growth hormone. Lab 505. Tannenbaum GS. Response: Effects of growth hormone-releasing factor
Invest 68: 584-591, 1993. in the brain. Science 232: 1272-1273, 1986.

732 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

506. Tannenbaum GS, Colle E, Wanamaker L, Gurd W, Goldman H, 530. Tolle V, Zizzari P, Tomasetto C, Rio MC, Epelbaum J, Bluet-Pajot MT.
Seemayer TA. Dynamic time-course studies of the spontaneously In vivo and in vitro effects of ghrelin/motilin-related peptide on growth
diabetic BB Wistar rat. II. Insulin-, glucagon-, and somatostatin-reactive hormone secretion in the rat. Neuroendocrinology 73: 54-61, 2001.
cells in the pancreas. Endocrinology 109: 1880-1887, 1981. 531. Tomasetto C, Karam SM, Ribieras S, Masson R, Lefèbvre O, Staub A,
507. Tannenbaum GS, Epelbaum J. Somatostatin. In: Kostyo JL, editor. Alexander G, Chenard MP, Rio MC. Identification and characteriza-
Handbook of Physiology. New York, Oxford: Oxford University Press tion of a novel gastric peptide hormone: The motilin-related peptide.
for the American Physiology Society, 1999, pp. 221-265. Gastroenterology 119: 395-405, 2000.
508. Tannenbaum GS, Epelbaum J, Bowers CY. Interrelationship between 532. Tranchand-Bunel D, Delbende C, Guy J, Jegou S, Jenks BJ, Mocaer
the novel peptide ghrelin and somatostatin/growth hormone-releasing E, Pelletier G, Vaudry H. Pro-opiomelanocortin neuronal systems. Rev
hormone in regulation of pulsatile growth hormone secretion. Neurol (Paris) 143: 471-489, 1987.
Endocrinology 144: 967-974, 2003. 533. Tschöp M, Smiley DL, Heiman ML. Ghrelin induces adiposity in
509. Tannenbaum GS, Epelbaum J, Colle E, Brazeau P, Martin JB. Anti- rodents. Nature 407: 908-913, 2000.
serum to somatostatin reverses starvation-induced inhibition of growth 534. Tsushima T, Irie M, Sakuma M. Radioimmunoassay for canine growth
hormone but not insulin secretion. Endocrinology 102: 1909-1914, hormone. Endocrinology 89: 685-693, 1971.
1978. 535. Tung E, Roberts CT, Heinemann GK, De Blasio MJ, Kind KL, van
510. Tannenbaum GS, Gurd W, Lapointe M. Leptin is a potent stimulator Wettere WH, Owens JA, Gatford KL. Increased placental nutrient trans-
of spontaneous pulsatile growth hormone (GH) secretion and the GH porter expression at midgestation after maternal growth hormone treat-
response to GH-releasing hormone. Endocrinology 139: 3871-3875, ment in pigs: A placental mechanism for increased fetal growth. Biol
1998. Reprod 87: 126, 2012.
511. Tannenbaum GS, Guyda HJ, Posner BI. Insulin-like growth factors: A 536. Tuomisto J, Mannisto P. Neurotransmitter regulation of anterior pitu-
role in growth hormone negative feedback and body weight regulation itary hormones. Pharmacol Rev 37: 249-332, 1985.
via brain. Science 220: 77-79, 1983. 537. Ulloa-Aguirre A, Blizzard RM, Garcia-Rubi E, Rogol AD, Link K,
512. Tannenbaum GS, Lapointe M, Beaudet A, Howard AD. Expression of Christie CM, Johnson ML, Veldhuis JD. Testosterone and oxandrolone,
growth hormone secretagogue-receptors by growth hormone-releasing a nonaromatizable androgen, specifically amplify the mass and rate of
hormone neurons in the mediobasal hypothalamus. Endocrinology 139: growth hormone (GH) secreted per burst without altering GH secretory
4420-4423, 1998. burst duration or frequency or the GH half-life. J Clin Endocrinol Metab
513. Tannenbaum GS, Ling N. The interrelationship of growth hormone 71: 846-854, 1990.
(GH)-releasing factor and somatostatin in generation of the ultradian 538. Vale W, Vaughan J, Yamamoto G, Spiess J, Rivier J. Effects of syn-
rhythm of GH secretion. Endocrinology 115: 1952-1957, 1984. thetic human pancreatic (tumor) GH releasing factor and somato-
514. Tannenbaum GS, Martin JB, Colle E. Ultradian growth hormone rhythm statin, triiodothyronine and dexamethasone on GH secretion in vitro.
in the rat: Effects of feeding, hyperglycemia, and insulin-induced hypo- Endocrinology 112: 1553-1555, 1983.
glycemia. Endocrinology 99: 720-727, 1976. 539. Van Cauter E, Kerkhofs M, Caufriez A, Van Onderbergen A, Thorner
515. Tannenbaum GS, Panerai AE, Friesen HG. Failure of beta-endorphin MO, Copinschi G. A quantitative estimation of growth hormone secre-
antiserum, naloxone, and naltrexone to alter physiologic growth hor- tion in normal man: Reproducibility and relation to sleep and time of
mone and insulin secretion. Life Sci 25: 1983-1990, 1979. day. J Clin Endocrinol Metab 74: 1441-1450, 1992.
516. Tannenbaum GS, van der Rest M, Downs TR, Frohman LA. Identifi- 540. Van Cauter E, Plat L, Copinschi G. Interrelations between sleep and
cation of a putative GH-releasing factor (GRF) batch as predominantly the somatotropic axis. Sleep 21: 553-566, 1998.
ovine CRF with a small quantity of human GRF. Endocrinology 118: 541. van den Berg G, Veldhuis JD, Frolich M, Roelfsema F. An amplitude-
1246-1248, 1986. specific divergence in the pulsatile mode of growth hormone (GH)
517. Tanner JM, Whitehouse RH, Marubini E, Resele LF. The adolescent secretion underlies the gender difference in mean GH concentrations in
growth spurt of boys and girls of the Harpenden growth study. Ann men and premenopausal women. J Clin Endocrinol Metab 81: 2460-
Hum Biol 3: 109-126, 1976. 2467, 1996.
518. Tanoh T, Shimatsu A, Ishikawa Y, Ihara C, Yanaihara N, Imura H. 542. van Essen M, Sundin A, Krenning EP, Kwekkeboom DJ. Neuroen-
Galanin-induced growth hormone secretion in conscious rats: Evidence docrine tumours: The role of imaging for diagnosis and therapy. Nat
for a possible involvement of somatostatin. J Neuroendocrinol 5: 183- Rev Endocrinol 10: 102-114, 2014.
187, 1993. 543. van Liempt S, Vermetten E, Lentjes E, Arends J, Westenberg H.
519. Tanriverdi F, Karaca Z, Unluhizarci K, Kelestimur F. Unusual effects Decreased nocturnal growth hormone secretion and sleep fragmen-
of GH deficiency in adults: A review about the effects of GH on skin, tation in combat-related posttraumatic stress disorder; potential pre-
sleep, and coagulation. Endocrine 47: 679-689, 2014. dictors of impaired memory consolidation. Psychoneuroendocrinology
520. Tatemoto K, Carlquist M, Mutt V. Neuropeptide Y–a novel brain pep- 36: 1361-1369, 2011.
tide with structural similarities to peptide YY and pancreatic polypep- 544. Vander Weele CM, Saenz C, Yao J, Correia SS, Goosens KA. Restora-
tide. Nature 296: 659-660, 1982. tion of hippocampal growth hormone reverses stress-induced hip-
521. Tatemoto K, Rokaeus A, Jornvall H, McDonald TJ, Mutt V. Galanin pocampal impairment. Front Behav Neurosci 7: 66, 2013.
- a novel biologically active peptide from porcine intestine. FEBS Lett 545. Veldhuis JD, Anderson SM, Patrie JT, Bowers CY. Estradiol supple-
164: 124-128, 1983. mentation in postmenopausal women doubles rebound-like release of
522. Tazearslan C, Huang J, Barzilai N, Suh Y. Impaired IGF1R signaling growth hormone (GH) triggered by sequential infusion and withdrawal
in cells expressing longevity-associated human IGF1R alleles. Aging of somatostatin: Evidence that estrogen facilitates endogenous GH-
Cell 10: 551-554, 2011. releasing hormone drive. J Clin Endocrinol Metab 89: 121-127, 2004.
523. Thakore JH, Dinan TG. Growth hormone secretion: The role of gluco- 546. Veldhuis JD, Anderson SM, Shah N, Bray M, Vick T, Gentili A, Mul-
corticoids. Life Sci 55: 1083-1099, 1994. ligan T, Johnson ML, Weltman A, Evans WS, Iranmanesh A. Neuro-
524. Theill LE, Karin M. Transcriptional control of GH expression and physiological regulation and target-tissue impact of the pulsatile mode
anterior pituitary development. Endocr Rev 14: 670-689, 1993. of growth hormone secretion in the human. Growth Horm IGF Res
525. Thomas GB, Cummins JT, Francis H, Sudbury AW, McCloud PI, 11(Suppl A): S25-37, 2001.
Clarke IJ. Effect of restricted feeding on the relationship between 547. Veldhuis JD, Evans WS, Bowers CY. Estradiol supplementation
hypophysial portal concentrations of growth hormone (GH)-releasing enhances submaximal feed-forward drive of growth hormone (GH)
factor and somatostatin, and jugular concentrations of GH in ovariec- secretion by recombinant human GH-releasing hormone-1,44-amide in
tomized ewes. Endocrinology 128: 1151-1158, 1991. a putatively somatostatin-withdrawn milieu. J Clin Endocrinol Metab
526. Thomas GB, Fairhall KM, Robinson IC. Activation of the hypothalamo- 88: 5484-5489, 2003.
pituitary-adrenal axis by the growth hormone (GH) secretagogue, 548. Veldhuis JD, Iranmanesh A, Ho KK, Waters MJ, Johnson ML,
GH-releasing peptide-6, in rats. Endocrinology 138: 1585-1591, Lizarralde G. Dual defects in pulsatile growth hormone secretion and
1997. clearance subserve the hyposomatotropism of obesity in man. J Clin
527. Thorner MO. Statement by the Growth Hormone Research Society on Endocrinol Metab 72: 51-59, 1991.
the GH/IGF-I axis in extending health span. J Gerontol A Biol Sci Med 549. Veldhuis JD, Reynolds GA, Iranmanesh A, Bowers CY. Twenty-four
Sci 64: 1039-1044, 2009. hour continuous ghrelin infusion augments physiologically pulsatile,
528. Tillet Y, Picard S, Bruneau G, Ciofi P, Wankowska M, Wojcik- nycthemeral, and entropic (feedback-regulated) modes of growth hor-
Gladysz A, Polkowska J. Hypothalamic arcuate neuropeptide Y- mone secretion. J Clin Endocrinol Metab 93: 3597-3603, 2008.
neurons decrease periventricular somatostatin-neuronal activity before 550. Veldhuis JD, Roelfsema F, Keenan DM, Pincus S. Gender, age, body
puberty in the female lamb: Morphological arguments. J Chem Neu- mass index, and IGF-I individually and jointly determine distinct GH
roanat 40: 265-271, 2010. dynamics: Analyses in one hundred healthy adults. J Clin Endocrinol
529. Tolle V, Bassant M-H, Zizzari P, Poindessous-Jazat F, Tomasetto C, Metab 96: 115-121, 2011.
Epelbaum J, Bluet-Pajot M-T. Ultradian rhythmicity of ghrelin secre- 551. Veldhuis JD, Roemmich JN, Richmond EJ, Bowers CY. Somatotropic
tion in relation with GH, feeding behavior, and sleep-wake patterns in and gonadotropic axes linkages in infancy, childhood, and the puberty-
rats. Endocrinology 143: 1353-1361, 2002. adult transition. Endocr Rev 27: 101-140, 2006.

Volume 6, April 2016 733


Growth Hormone Axis Comprehensive Physiology

552. Veldhuis JD, Roemmich JN, Rogol AD. Gender and sexual maturation- 574. Wiedemann E, Schwartz E, Frantz AG. Acute and chronic estrogen
dependent contrasts in the neuroregulation of growth hormone secretion effects upon serum somatomedin activity, growth hormone, and pro-
in prepubertal and late adolescent males and females–a general clinical lactin in man. J Clin Endocrinol Metab 42: 942-952, 1976.
research center-based study. J Clin Endocrinol Metab 85: 2385-2394, 575. Willesen MG, Kristensen P, Rømer J. Co-localization of growth hor-
2000. mone secretagogue receptor and NPY mRNA in the arcuate nucleus of
553. Vgontzas AN, Mastorakos G, Bixler EO, Kales A, Gold PW, Chrousos the rat. Neuroendocrinology 70: 306-316, 1999.
GP. Sleep deprivation effects on the activity of the hypothalamic- 576. Willoughby JO, Brogan M, Kapoor R. Hypothalamic interconnections
pituitary-adrenal and growth axes: Potential clinical implications. Clin of somatostatin and growth hormone releasing factor neurons. Neu-
Endocrinol (Oxf) 51: 205-215, 1999. roendocrinology 50: 584-591, 1989.
554. Vidal S, Horvath E, Kovacs K, Lloyd RV, Smyth HS. Reversible transd- 577. Willoughby JO, Brogan M, Kapoor R. Intrahypothalamic actions of
ifferentiation: Interconversion of somatotrophs and lactotrophs in pitu- somatostatin and growth hormone releasing factor on growth hormone
itary hyperplasia. Mod Pathol 14: 20-28, 2001. secretion. Neuroendocrinology 50: 592-596, 1989.
555. Vijayakumar A, Novosyadlyy R, Wu Y, Yakar S, LeRoith D. Biolog- 578. Willoughby JO, Kapoor R, Mackenzie L. Intrahypothalamic mu-, not
ical effects of growth hormone on carbohydrate and lipid metabolism. delta- or kappa-opioid receptor activation causes growth hormone secre-
Growth Horm IGF Res 20: 1-7, 2010. tion. J Neuroendocrinol 3: 149-154, 1991.
556. Vijayan E, Krulich L, McCann SM. Stimulation of growth hormone 579. Wilson EE, Word RA, Byrd W, Carr BR. Effect of superovulation
release by intraventricular administration of 5HT or quipazine in with human menopausal gonadotropins on growth hormone levels in
unanesthetized male rats. Proc Soc Exp Biol Med 159: 210-212, 1978. women. J Clin Endocrinol Metab 73: 511-515, 1991.
557. Vitiello MV, Schwartz RS, Moe KE, Mazzoni G, Merriam GR. Treating 580. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S,
age-related changes in somatotrophic hormones, sleep, and cognition. Zhang Y, Bernal D, Pons S, Shulman GI, Bonner-Weir S, White MF.
Dialogues Clin Neurosci 3: 229-236, 2001. Disruption of IRS-2 causes type 2 diabetes in mice. Nature 391: 900-
558. Wagner C, Caplan SR, Tannenbaum GS. Genesis of the ultradian 904, 1998.
rhythm of GH secretion: A new model unifying experimental observa- 581. Wolkow CA, Kimura KD, Lee MS, Ruvkun G. Regulation of C. elegans
tions in rats. Am J Physiol 275: E1046-1054, 1998. life-span by insulinlike signaling in the nervous system. Science 290:
559. Wakabayashi I, Demura R, Miki N, Ohmura E, Miyoshi H, Shizume K. 147-150, 2000.
Failure of naloxone to influence plasma growth hormone, prolactin, and 582. Wren AM, Small CJ, Fribbens CV, Neary NM, Ward HL, Seal
cortisol secretions induced by insulin hypoglycemia. J Clin Endocrinol LJ, Ghatei MA, Bloom SR. The hypothalamic mechanisms of the
Metab 50: 597-599, 1980. hypophysiotropic action of ghrelin. Neuroendocrinology 76: 316-324,
560. Wang H-J, Geller F, Dempfle A, Schäuble N, Friedel S, Lichtner P, 2002.
Fontenla-Horro F, Wudy S, Hagemann S, Gortner L, Huse K, Rem- 583. Wynick D, Smith DM, Ghatei M, Akinsanya K, Bhogal R, Purkiss P,
schmidt H, Bettecken T, Meitinger T, Schäfer H, Hebebrand J, Hinney Byfield P, Yanaihara N, Bloom SR. Characterization of a high-affinity
A. Ghrelin receptor gene: Identification of several sequence variants in galanin receptor in the rat anterior pituitary: Absence of biological effect
extremely obese children and adolescents, healthy normal-weight and and reduced membrane binding of the antagonist M15 differentiate it
underweight students, and children with short normal stature. J Clin from the brain/gut receptor. Proc Natl Acad Sci U S A 90: 4231-4235,
Endocrinol Metab 89: 157-162, 2004. 1993.
561. Watanobe H, Habu S. Leptin regulates growth hormone-releasing fac- 584. Xie TY, Ngo ST, Veldhuis JD, Jeffery PL, Chopin LK, Tschöp M,
tor, somatostatin, and alpha-melanocyte-stimulating hormone but not Waters MJ, Tolle V, Epelbaum J, Chen C, Steyn FJ. Effect of deletion
neuropeptide Y release in rat hypothalamus in vivo: Relation with of ghrelin-o-acyltransferase on the pulsatile release of growth hormone
growth hormone secretion. J Neurosci 22: 6265-6271, 2002. in mice. J Neuroendocrinol. 2015 Oct 7. doi: 10.1111/jne.12327. [Epub
562. Wehrenberg WB, Baird A, Klepper R, Mormede P, Ling N. Interactions ahead of print]
between growth hormone-releasing hormone and glucocorticoids in 585. Xu J, Gontier G, Chaker Z, Lacube P, Dupont J, Holzenberger
male rats. Regul Pept 25: 147-155, 1989. M. Longevity effect of IGF-1R(+/−) mutation depends on genetic
563. Wehrenberg WB, Bloch B, Ling N. Pituitary secretion of growth hor- background-specific receptor activation. Aging Cell 13: 19-28, 2014.
mone in response to opioid peptides and opiates is mediated through 586. Yagi H, Kaji H, Sato M, Okimura Y, Chihara K. Effect of intravenous
growth hormone-releasing factor. Neuroendocrinology 41: 13-16, 1985. or intracerebroventricular injections of His-D-Trp-Ala-Trp-D-Phe-Lys-
564. Wehrenberg WB, Giustina A. Feedback regulation of growth hormone NH2 on GH release in conscious, freely moving male rats. Neuroen-
secretion. In: Kostyo JL, editor. Handbook of Physiology. New York docrinology 63: 198-206, 1996.
Oxford: Oxford University Press for the American Physiology Society, 587. Yamasaki H, Prager D, Gebremedhin S, Melmed S. Insulin-like growth
1999, pp. 299-327. factor-I (IGF-I) attenuation of growth hormone is enhanced by over-
565. Wehrenberg WB, Janowski BA, Piering AW, Culler F, Jones KL. Gluco- expression of pituitary IGF-I receptors. Mol Endocrinol 5: 890-896,
corticoids: Potent inhibitors and stimulators of growth hormone secre- 1991.
tion. Endocrinology 126: 3200-3203, 1990. 588. Yamashita S, Melmed S. Effects of insulin on rat anterior pituitary cells.
566. Wehrenberg WB, Wiviott SD, Voltz DM, Giustina A. Pyridostigmine- Inhibition of growth hormone secretion and mRNA levels. Diabetes 35:
mediated growth hormone release: Evidence for somatostatin involve- 440-447, 1986.
ment. Endocrinology 130: 1445-1450, 1992. 589. Yamashita S, Melmed S. Insulin regulation of rat growth hormone gene
567. Weibel L, Follenius M, Spiegel K, Gronfier C, Brandenberger G. transcription. J Clin Invest 78: 1008-1014, 1986.
Growth hormone secretion in night workers. Chronobiol Int 14: 49- 590. Yamashita S, Ong J, Melmed S. Regulation of human growth hormone
60, 1997. gene expression by insulin-like growth factor I in transfected cells.
568. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper J Biol Chem 262: 13254-13257, 1987.
JL. Effect of fasting, refeeding, and dietary fat restriction on plasma 591. Yamauchi N, Shibasaki T, Ling N, Demura H. In vitro release of growth
leptin levels. J Clin Endocrinol Metab 82: 561-565, 1997. hormone-releasing factor (GRF) from the hypothalamus: Somatostatin
569. Weissberger AJ, Ho KK, Lazarus L. Contrasting effects of oral and inhibits GRF release. Regul Pept 33: 71-78, 1991.
transdermal routes of estrogen replacement therapy on 24-hour growth 592. Yang J, Brown MS, Liang G, Grishin NV, Goldstein JL. Identification
hormone (GH) secretion, insulin-like growth factor I, and GH-binding of the acyltransferase that octanoylates ghrelin, an appetite-stimulating
protein in postmenopausal women. J Clin Endocrinol Metab 72: 374- peptide hormone. Cell 132: 387-396, 2008.
381, 1991. 593. Yokote R, Sato M, Matsubara S, Ohye H, Niimi M, Murao K, Takahara
570. Weltman A, Weltman JY, Hartman ML, Abbott RD, Rogol AD, Evans J. Molecular cloning and gene expression of growth hormone-releasing
WS, Veldhuis JD. Relationship between age, percentage body fat, fit- peptide receptor in rat tissues. Peptides 19: 15-20, 1998.
ness, and 24-hour growth hormone release in healthy young adults: 594. Yulyaningsih E, Loh K, Lin S, Lau J, Zhang L, Shi Y, Berning BA,
Effects of gender. J Clin Endocrinol Metab 78: 543-548, 1994. Enriquez R, Driessler F, Macia L, Khor EC, Qi Y, Baldock P, Sainsbury
571. Wennink JM, Delemarre-van de Waal HA, Schoemaker R, Blaauw G, A, Herzog H. Pancreatic polypeptide controls energy homeostasis via
van den Braken C, Schoemaker J. Growth hormone secretion patterns Npy6r signaling in the suprachiasmatic nucleus in mice. Cell Metab
in relation to LH and estradiol secretion throughout normal female 19: 58-72, 2014.
puberty. Acta endocrinologica 124: 129-135, 1991. 595. Zhang J, Obal F, Jr., Fang J, Collins BJ, Krueger JM. Non-rapid eye
572. Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, movement sleep is suppressed in transgenic mice with a deficiency in
Mendelsohn FA. Localization and characterization of insulin receptors the somatotropic system. Neurosci Lett 220: 97-100, 1996.
in rat brain and pituitary gland using in vitro autoradiography and 596. Zhang J, Obal F, Jr., Zheng T, Fang J, Taishi P, Krueger JM. Intrapre-
computerized densitometry. Endocrinology 121: 1562-1570, 1987. optic microinjection of GHRH or its antagonist alters sleep in rats.
573. Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Mendel- J Neurosci 19: 2187-2194, 1999.
sohn FA. Localization and characterization of insulin-like growth 597. Zhang JV, Ren P-G, Avsian-Kretchmer O, Luo C-W, Rauch R,
factor-I receptors in rat brain and pituitary gland using in vitro autora- Klein C, Hsueh AJW. Obestatin, a peptide encoded by the ghrelin
diography and computerized densitometry∗ a distinct distribution from gene, opposes ghrelin’s effects on food intake. Science 310: 996-999,
insulin receptors. J Neuroendocrinol 1: 369-377, 1989. 2005.

734 Volume 6, April 2016


Comprehensive Physiology Growth Hormone Axis

598. Zhao T-J, Liang G, Li RL, Xie X, Sleeman MW, Murphy AJ, Valen- 600. Zizzari P, Halem H, Taylor J, Dong JZ, Datta R, Culler MD, Epelbaum
zuela DM, Yancopoulos GD, Goldstein JL, Brown MS. Ghrelin O- J, Bluet-Pajot MT. Endogenous ghrelin regulates episodic growth hor-
acyltransferase (GOAT) is essential for growth hormone-mediated sur- mone (GH) secretion by amplifying GH Pulse amplitude: Evidence
vival of calorie-restricted mice. Proc Natl Acad Sci USA 107: 7467- from antagonism of the GH secretagogue-R1a receptor. Endocrinology
7472, 2010. 146: 3836-3842, 2005.
599. Zhou Y, Rui L. Leptin signaling and leptin resistance. Front Med 7: 601. Zizzari P, Longchamps R, Epelbaum J, Bluet-Pajot MT. Obestatin
207-222, 2013. partially affects ghrelin stimulation of food intake and growth hormone
secretion in rodents. Endocrinology 148: 1648-1653, 2007.

Volume 6, April 2016 735


View publication stats

You might also like