ChapterPDF Available

Neuroendocrine and Metabolic Regulation of Plasma Growth Hormone Secretory Profiles

Authors:

Abstract and Figures

Like many other neuroendocrine hormones, growth hormone (GH; somatotropin) secretion is pulsatile with regular releasing bursts on a relatively low constitutive basal secretion. This chapter discusses current knowledge of the regulation and the function of GH pulsatile profiles, with new development of laboratory approaches and introduces knowledge about GH functions on metabolic regulation in addition to the conventional concept of a role in regulating body growth. As reported in the literature, amplitude, frequency, and regularity of GH secretion are tightly linked to metabolic conditions with clear species and gender differences. In response to negative and positive energy balances, the GH pulsatile pattern changes to mobilize or store energy in adipose, muscle, and liver in order to accommodate the changing nutritional conditions. Changes in GH pulsatility are achieved through regulating the hypothalamo–pituitary GH axis with altered levels of key stimulatory and inhibitory hormones, GH-releasing hormone (GHRH) and somatostatin (SRIF, somatotropin release inhibiting factor). The hypothalamic–pituitary GH axis is constantly under the influence of peripheral metabolic factors, such as lipid and glucose levels; peripheral metabolic regulatory hormones, such as leptin and insulin; and central metabolic regulatory neuropeptides, such as neuropeptide Y and melanocortin. Regulation of the hypothalamic–pituitary GH axis is achieved through activation of cell membrane receptors, intracellular signaling pathways, and membrane ion channels. Detailed regulatory mechanisms are discussed in this chapter in order to understand the coupling of cell electrophysiological properties and the hormone secretory process of exocytosis in hypothalamic neurons and pituitary GH-secreting somatotrophs. Technical advances in electrophysiology, cell imaging analysis, and real-time in vivo hormone analysis are discussed to deepen the understanding of physiological and pathophysiological regulation of GH secretion. Future directions are also discussed, as are unanswered questions in this field.
Content may be subject to copyright.
Review
Insulin and Growth Hormone Balance:
Implications for Obesity
Zhengxiang Huang,
1
Lili Huang,
1
Michael J. Waters,
2
and Chen Chen
1,
*
Disruption of endocrine hormonal balance (i.e., increased levels of insulin, and
reduced levels of growth hormone, GH) often occurs in preobesity and obesity.
Using distinct intracellular signaling pathways to control cell and body metabo-
lism, GH and insulin also regulate each others secretion to maintain overall
metabolic homeostasis. Therefore, a comprehensive understanding of insulin
and GH balance is essential for understanding endocrine hormonal contributions
to energy storage and utilization. In this review we summarize the actions of, and
interactions between, insulin and GH at the cellular level, and highlight the associ-
ation between the insulin/GH ratio and energy metabolism, as well as fat accumu-
lation. Use of the [insulin]:[GH] ratio as a biomarker for predictingthe development
of obesity is proposed.
InsulinGH balance
Obesity (see Glossary) is a major health issue in the contemporary world and has a strong
association with type 2 diabetes (T2D), cardiovascular disease, and cancer. Hormonal imbalance
often occurs before or during obesity and may play a causal role in the development of obesity
and associated chronic diseases. Insulin and GH, two vital counter-regulatory hormones for glu-
cose and lipid metabolism, are often dysregulated in obesity (increased insulin and reduced GH)
[1], although the cause and consequence of such hormonal imbalance have not been fully clari-
ed. In the evolutionary view, insulin promotes energy storage in the condition of energy surplus,
whereas GH promotes lipid mobilization and oxidation when food is sparse [2]. During natural
selection over the past million years, the human body has evolved an endocrine system adapted
to an environment with insufcient food supply. However, in modern developed societies with
easy food access, overnutrition often occurs. This shifts the dominant role towards insulin
(increased) and away from GH (suppressed) [3,4]. This shift hinders lipid breakdown and pro-
motes further energy storage and lipid synthesis, which exacerbates the development of obesity.
Surprisingly, a comprehensive review of the balance between insulinand GH (insulinGH balance)
and its physiological signicance is lacking. In this review we give a brief summary of the signaling
crosstalk between insulin and GH at the cellular level, and summarize recent ndings. We analyze
changes in the insulinGH balance and the association with energy metabolism in various
physiological and pathophysiological conditions, and we discuss the implications of the insulin
GH balance in the treatment of obesity.
Physiological Roles of Insulin and GH
Insulin and GH both display pulsatile secretion. Synthesized in pancreatic βcells, peak
secretion of insulin is predominant after each meal with small oscillations throughout the day
[5]. GH is released from somatotrophs in the pituitary gland,anditssecretionpatternis
characterized by low baseline secretion with several dominant pulses at rhythmic intervals
[6]. Although the physiological impact of pulsatile secretion is not fully understood, pulsatility
may contribute to receptor mobilization, turnover, and associated cellular signaling, including
sexual dimorphism [7].
Highlights
Insulin and GH are counter-regulatory
hormones in terms of glucose and lipid
metabolism, but act synergistically in
protein metabolism. They also mutually
regulate the secretion of each other,
forming a complex regulatory network.
The balance between insulin and GH is
associated with substrate and energy
metabolism. In obesity, the hormonal im-
balance (high insulin and low GH) pro-
motes further fat accumulation.
Clinical data from various physiological
and pathophysiological conditions with
insulin and GH changes indicate that
the [insulin]:[GH] ratio correlates nega-
tively with energy expenditure and corre-
lates positively with fat accumulation.
The [insulin]:[GH] ratio may serve as a
biomarker for monitoring and predicting
the development of obesity. Modulation
of insulinGH balance is a promising tar-
get for managing obesity.
1
School of Biomedical Sciences,
University of Queensland, St Lucia,
Brisbane, Australia
2
Institute for Molecular Bioscience,
University of Queensland, St Lucia,
Brisbane, Australia
*Correspondence:
chen.chen@uq.edu.au (C. Chen).
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx https://doi.org/10.1016/j.tem.2020.04.005 1
© 2020 Elsevier Ltd. All rights reserved.
Trends in Endocrinology & Metabolism
TEM 1513 No. of Pages 13
Intracellular Signaling Pathways of Insulin and GH
The classic insulin receptor signaling pathways generally include PI3K/Akt and Ras/MAPK, which
regulate metabolic and mitogenic effects, respectively. Activation of the PI3K/Akt pathway is
dependent on tyrosine phosphorylation of the adaptors insulin receptor substrate 1 and
2 (IRS1/2), whereas Ras/MAPK can also be activated through IRS1/2-independent pathways
[8]. The mammalian target of rapamycin complex 1 (mTORC1), that is activated by Akt, is an
essential component in the regulation of lipid and protein metabolism [8].
The GH receptor is a member of the class I cytokine receptor family. Following GH binding, it
activates the Janus kinase 2 (JAK2)signal transducer and activator of transcription (STAT) and
Src/MAPK pathways [9], where the former dominantly regulates metabolic effects and the latter
regulates mitogenic function. JAK2 controls metabolic effects by activating STAT1, 3, 5a, and
5b, of which STAT5b is the most prominent and also promotes insulin-like growth factor 1
(IGF-1) production, hence linear growth [10]. Some studies suggest that JAK2 also phosphory-
lates IRS1/2 and activates the PI3K/Akt pathway [1113]. However, these studies either used
a supraphysiological GH dose [12,13] or did not determine the physiological effects of GH
administration [11], and their conclusions may not apply to a physiological scenario.
Because both insulin and GH regulate lipid/glucose/protein metabolism, their effects and
signaling pathways are further discussed based on target substrate metabolism. We limit
coverage of insulin receptor signaling because this has been thoroughly reviewed [14]. The
actions and signaling pathways of insulin and GH are summarized in Figure 1.
Actions of Insulin and GH Receptor Signaling on Substrate Metabolism
Lipid Metabolism
As a fat-sparing hormone, insulin promotes lipogenesis while inhibiting lipolysis and lipid oxidation
(Figure 1). By contrast, the major effect of GH on lipid metabolism is through inducing lipolysis [2],
which increases free fatty acid (FFA) and glycerol release from adipose tissue into the circulation.
The mechanism of increased lipolysis by GH is not yet fully understood. Some studies suggest
that GH increases the expression and action of β-adrenergic receptors [15,16], which activate
hormone-sensitive lipase (HSL) by increasing the intracellular level of cAMP ([cAMP]i) via activation
of protein kinase A (PKA) [17]. Other studies suggest that GH increases lipolysis through activa-
tion of MEK/ERK and inhibition of PPARγand fat-specic protein 27 (FSP27) [18]. Although
increased circulating FFA levels are associated with metabolic diseases (e.g., non-alcoholic
fatty liver disease and T2D) [19], physiologically increasing GH secretion (e.g., exercise or fasting)
does not lead to such diseases. This paradox can be partially explained by GH-induced lipid
oxidation. GH increases brown adipose tissue (BAT) function and white adipose tissue (WAT)
browning/beiging [16] (discussed later). GH also stimulates lipid uptake in muscle by increasing
the activity of muscle lipoprotein lipase (LPL) [20]. FFA released from WAT is taken up and
oxidized in other tissues, and the net effect of elevated GH therefore promotes reduction of
whole-body fat content. Recent studies have also found that GHRJAK2STAT5 signaling in-
hibits lipid uptake and de novo lipogenesis in the liver, partly through inhibition of PPARγand
downstream CD36 [2124], indicating that GH has a direct effect on hepatic lipid metabolism
and can reduce non-alcoholic fatty liver disease [23].
Glucose Metabolism
Upon food intake, secreted insulin promotes glucose uptake and glycogen synthesis, while
inhibiting gluconeogenesis through the PI3K/Akt pathway (Figure 1). GH stimulates gluconeogen-
esis [2] in the liver, partly through the STAT5 pathway, which includes an increase in phosphoenol-
pyruvate carboxykinase (PEPCK), glucose 6-phosphatase [25], and pyruvate dehydrogenase
Glossary
Acromegaly: a condition in which the
pituitary produces excessive GH during
adulthood, mostly due to a pituitary
adenoma.
Growth hormone (GH) deficiency:
the pituitary is unable to produce
sufcient GH because of primary lesion.
Childhood-onset GH deciency is mainly
due to genetic defects, whereas adult
GH deciencycan be caused by multiple
factors including trauma, infections,
tumors, or radiation therapy. Reduced
secretion of GH in obesity does not
belong to this category in this review.
However, it is often referred as 'relative
GH deciency' in obesity in most
literature.
Insulin-like growth factor 1 (IGF-1):
IGF-1 is produced by a variety of cell
types following GH stimulation.
Circulating IGF-1 is mainly produced by
the liver and acts as a negative regulator
of GH secretion. Local IGF-1 produced
by bone and musclecontributes to linear
growth and protein anabolism.
Obesity: according to the World Health
Organization, obesity is dened as a
body mass index (BMI, body weight in
kg divided by height in m squared) of 30
or more.
Pegvisomant: a drug used for treating
acromegaly in the clinic that blocks
downstream signaling of the GH
receptor. It is a modied version of
human GH that competes with native
GH for the GH receptor and prevents its
activation.
Pituitary gland: an important
endocrine organ located in the brain,
under the h ypothalam us. It res ponds to
hormonalsignals from the hypothalamus
(e.g., GH-releasing hormone) and
produces hormones (e.g., GH) to
regulate body functions.
Sulfonylurea: an antidiabetic drug that
increases insulin secretion by closing
ATP-sensitive potassium channels on
pancreatic βcells.
Trends in Endocrinology & Metabolism
2Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
kinase 4 (PDK4) gene expression [26]. GH also stimulates glycogenolysis in the liver [27]. A recent
study nds that decreased debranching enzyme (AGL) and increased glycogen branching enzyme
(GBE1) expression are direct STAT5 targets in the liver of mice with disruptedSTAT5 signaling [24],
suggesting an important role for AGL and GBE1 in GH-induced glycogenolysis.
Protein Metabolism
Although insulin and GH have distinct effects on lipid and glucose metabolism, both promote
protein anabolism by increasing protein synthesis and reducing protein breakdown. Further,
both insulin and IGF-1 promote protein anabolism through mTORC1, which is activated by
PI3K/Akt [28].GHhasbeenshowntopromoteproteinsynthesisthroughthesamepathway
[29], but it seems that this effect is mainly through IGF-1 autocrine/paracrine action because
Insulin/IGF-1 receptor
GH receptor
Src
MAPK/
ERK
Ras
PI3K/
Akt
STAT5
IGF-1 SREBP GSK3 FOXO1
mTORC1
Protein
synthesis
PPARγPPARα
UCP1PPARγ CD36 PEPCK/
PDK4 HSL
Proliferaon
Differenaon
Development
Lipid ulizaon ↓
Lipogenesis ↑
Lipolysis ↓
Glycogen synthesis ↑
Gluconeogenesis ↓
GLUT4 translocaon ↑
JAK2
PPARγ
β-adrenergic
receptors
[cAMP]
HSL
Lipolysis ↑
Lipogenesis ↓
IRS
Insulin
[cAMP]
IGF-1
GH
Lipogenesis ↓
TG uptake (liver) ↓
Brown/beige fat funcon ↑
Gluconeogenesis ↑
Glycogenolysis ↑
AGL
Trends
Trends
in
in
Endocrinology
Endocrinology &
Metabolism
Metabolism
Figure 1. Signaling Pathways and Their Effectson Substrate Metabolism of Insulin and Growth Hormone(GH)
Receptors. The major signaling pathways downstream of the insulin receptor are PI3K/Akt and Ras/MAPK,and the primary
signaling pathways of the GH receptor are JAK2/STAT and Src/MAPK. While PI3K/Akt and JAK2/STAT pathways contribute
to the metabolic effects of insulin and GH receptor signaling, the MAPK pathway is responsible for mitogenic effects. The
distinct effects in glucose and lipid metabolism of insulin and GH are shown in yellow and red, respectively. The synergistic
effects of insulin and GH are shown in green. The dashed line indicates that the physiological effects of JAK2-PI3K/Akt
pathway are not clear. Abbreviations: AGL, glycogen debranching enzyme; FOXO1, Forkhead Box O1; GLUT4, glucose
transporter type 4; GNG, gluconeogenesis; GSK3, glycogen synthase kinase 3; HSL, hormone-sensitive lipase; IRS,
insulin receptor substrate; JAK2, Janus kinase 2; mTORC1, mammalian target of rapamycin complex 1; PDK4, pyruvate
dehydrogenase k inase 4; PEPCK, phosphoenolpyruvate carboxykinase; PI3K/Akt, p hosphatidylin ositol 3-kinase/protein
kinase B; PPAR, peroxisome proliferator-activated receptor; SREBP, sterol regulatory element-binding protein; STAT,
signal transducer and activator of transcription; TG, triglyceride; UCP1, uncoupling protein 1.
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 3
GH administration increases muscle mass in wild-type mice but not in mice lacking the IGF-1 re-
ceptor specically in muscle [30]. In addition, GH increases lipolysis and FFA levels in stress states
(such as fasting and exercise), thus reducing protein usage and exerting a protein-sparing effect [31].
Interactions between Insulin and GH
Molecular and Cellular Effects
Although exerting similar effects on protein metabolism, insulin and GH show opposite effects
on glucose and lipid metabolism. As discussed earlier, insulin reduces blood glucose and
promotes lipogenesis, whereas GH increases blood glucose and facilitates lipolysis and lipid
oxidation. This antagonism is further evidenced by insulin resistance following GH administra-
tion or in acromegaly patients [32]; downregulation of GH receptor signaling is found in vitro
following insulin pretreatment [33]. In addition to opposing effects on the same gene target
(e.g., insulin inhibits HSL, whereas GH activates HSL), GH antagonizes insulin action through
post-receptor signaling pathways. Further, the insulin sensitivity seen in GH receptor-deleted
mice is partly a consequence of the removal of suppression of insulin-sensitizing hormones
such as adiponectin, FGF1, and IL-15 [24].
At the post-receptor level, GH transgenic mice show decreased insulin receptor levels and
decreased insulin-stimulated phosphorylation of crucial molecules in the insulin receptor
pathway, including insulin receptor, IRS1/2, and PI3K [34]. In addition, GH induces p85α
subunit, a regulatory subunit of PI3K [35], and thus inhibits PI3K activity via p110 catalytic
subunit inhibition [35] as well as by phosphatase and tensin homolog (PTEN) activation [36]. GH
also activates suppressor of cytokine signaling (SOCS) 1 and3, leading to insulin signaling inhibition
[37]. These ndings demonstrate the antagonizing effect of GH on insulin action at the cellular level.
Further, although physiological insulin is necessary for the maintenance of surface GH receptors,
chronic high insulin levels reduce surface GH receptor availability [38] and GH-induced phosphor-
ylation of JAK2 and STAT5b in vitro [34], indicating that hyperinsulinemia may lead to GH resis-
tance. The intracellular interactions between insulin and GH are summarized in Figure 2.
Reciprocal Regulation of Secretion
The regulation of GH secretion by insulin is complex. An acute increase in insulin levels by exog-
enous injection, which depresses blood glucose levels below the normal range, stimulates GH
secretion. This is seen in the insulin tolerance test (Figure 3A), a widely accepted method for eval-
uating GH secretion capacity in adults [39]. However, there is no evidence that a physiological
increase in insulin levels (i.e., glucose-stimulated insulin secretion, GSIS) (Figure 3B) inuences
GH secretion. Conversely, chronic hyperinsulinemia, that is often seen in obesity and T2D
(Figure 3C), can directly reduce GH secretion in somatotrophs independently of IGF-1 signaling
[40]. In addition, hyperinsulinemia reduces IGFBP-1 production in the liver [41], which in-
creases circulating free IGF-1 and thus inhibits GH secretion by IGF-1 negative feedback on
the pituitary gland. Conversely, low insulin levels often lead to increased GH secretion. This is
mainly due to reduced surface expression of the GH receptor in the liver [38], leading to re-
duced IGF-1 production, which diminishes the IGF-1 negative feedback on the pituitary
gland. This is often observed in type 1 diabetes (T1D) patients and in the fasting state in
humans (Figure 3D,E).
Likewise, the regulation of insulin secretion by GH is also sophisticated. High levels of circulat-
ing GH in acromegaly patients (Figure 3F) cause hyperinsulinemia because of the insulin-
antagonistic effect of GH, discussed above. Low GH levels in the short term, as seen in
childhood-onset growth hormone deficiency adolescents (Figure 3G), increase insulin sen-
sitivity [42], probably due to the removal of the insulin-antagonistic effect of GH. In the long
Trends in Endocrinology & Metabolism
4Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
term, adult GH-decient patients (Figure 3H) show similar fasting hyperinsulinemia to body
mass index (BMI)-matched obese individuals, probably due to insulin resistance [43] caused
by reduced fat breakdown and increased fat mass. Apart from distinct insulin sensitivity in
short- and long-term GH deciency, it is generally acknowledged that GH is required for pan-
creatic βcell proliferation [44]. Thus, children with isolated-GH deciency are often reported to
have reduced βcell function [45].
The Ratio of Circulating Insulin to GH Correlates with Energy Expenditure and Fat
Accumulation
The overall analysis of insulin and GH in various physiological and pathophysiological conditions
(Table 1) supports the use of the ratio of circulating insulin to GH ([insulin]:[GH] ratio) to reect en-
ergy metabolism. To be specic, the higher the [insulin]:[GH] ratio, the lower the energy expendi-
ture, the more fat accumulation, and vice versa. Details are discussed in the following sections.
Although analysis of insulin in both postabsorptive and postprandial states as a function of pulsa-
tile GH components (i.e., pulsatile, basal, GH mass per pulse) may provide a more comprehen-
sive and more in-depth understanding of the role of the hormone properties, it should be noted
that the overall levels of insulin and GH mentioned here are restricted to fasting insulin and total
GH secretion, respectively, owing to the limited number of parameters assessed in most studies.
Conditions with Decreased [Insulin]:[GH] Ratio
Reduced [insulin]:[GH] ratio is found in T1D patients [46], individuals performing aerobic exercise
[47,48], and acromegaly patients, who have ~10-fold increased basal GH secretion [49,50] with a
compensatory twofold increase in fasting insulin level [49](Table 1, upper panel). As expected,
Insulin receptor
GH receptor
IRS
PI3K
STAT5
JAK2
SOCSs
p110
p85
PTEN
JAK2
IRS
GH
Insulin
?
?
Trends
Trends
in
in
Endocrinology
Endocrinology &
Metabolism
Metabolism
Figure 2. Intracellular Interaction between Insulin and Growth Hormone (GH). Downstream signaling by the insulin
receptor (yellow) and the GH receptor (red) antagonizes each other at the cellular level through various pathways. The
question mark indicates an unknown mechanism. Abbreviations: PTEN, phosphatase and tensin homolog; SOCS,
suppressor of cytokine signaling.
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 5
T1D patients [51,52], regularly exercised individuals [53], and acromegaly patients [50,54] show
decreased fat mass compared with body weight-matched controls. In terms of energy
expenditure, studies show that T1D patients have increased basal energy expenditure both
untreated and after the withdrawal of insulin treatment [55,56]. Exercising individuals show higher
basal metabolic rate both in self-comparison trials [57,58] and relative to non-exercise individuals
[59]. Several studies also show that acromegaly patients have increased energy expenditure
[49,50,54,60,61]. Therefore, reduced [insulin]:[GH] ratio seems to be associated with reduced
Table 1. Changes in Energy Expenditure and Fat Mass in Different Diseases/Conditions with Increased or
Decreased [Insulin]:[GH] Ratio
Condition Insulin GH Energy
expenditure
Fat
mass
Refs
Decreased [insulin]:
[GH] ratio
T1D ↓↑↑ ↓[46,51,52,55,56]
Exercise ↓↑↑ ↓[47,48,53,5759]
Acromegaly ↑↑↑ ↓[49,50,54,60,61]
Increased [insulin]:
[GH] ratio
AGHD ↑↓or ↓↑[43,73,80]
Sleep
disorders
or
↓↓ [64,65,67,69,70,7578,82]
Aging or
↓↓ [62,68,71,72]
Obese
PCOS
↑↓↓ ↑[63,66,74,79]
Simple
obesity
↑↓↓ ↑[83]
High insulin
Low insulin
(T1D)
Insulin
GHR on liver ↓ IGF-1 ↓
GH
High GH
(acromegaly)
Low GH
(GHD)
Glucose ↓
Acute
Chronic
(obesity)
Insulin resistance
(ITT)
GSIS doesn't
change GH
(GSIS)
Pancreac
β cells
Somatotrophs
Increased fat mass
Insulin resistance
Childhood
Adult
+
-
-
+
+
Conditions Primary
change
Secondary
change
(A)
ITT Insulin ↑ GH ↑
(B)
GSIS Insulin ↑ GH ↔
(C) Obesity or T2D
Insulin ↑ GH ↓
(D)
T1D Insulin ↓ GH ↑
(E)
Fasting Insulin ↓ GH ↑
(F)
Acromegaly GH ↑↑ Insulin ↑
(G) Childhood GHD
GH ↓ Insulin ↓
(H) Adult GHD GH ↓ Insulin ↑
+
Trends
Trends
in
in
Endocrinology
Endocrinology &
Metabolism
Metabolism
Figure 3. Inter-regulation between Insulin and Growth Hormone (GH). Primary increase in insulin secretion leads to either increased (A), unchanged (B), or
decreased (C) GH secretion, according to the conditions. Primary decrease in insulin secretion leads to increased GH secretion (D,E). Primary increase in GH secretion
leads to increased insulin secretion by compensation (F), whereas primary decrease in GH secretion (i.e., GH deciency) leads to either decreased (G) or increased (H)
insulin secretion as a function of onset age. Examples of diseases/conditions for changed insulin or GH secretion are indicated in the brackets and table. Symbols and
abbreviations: , increase; , decrease; , no change; GHD, growth hormone deciency; GHR, growth hormone receptor; GSIS, glucose-stimulated insulin secretion;
ITT, insulin tolerance test, a widely accepted method for evaluating GH secretion capacity in humans; T1D/T2D, type 1 and 2 diabetes.
Trends in Endocrinology & Metabolism
6Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
fat mass and increased energy expenditure, at least in untreated T1D patients, regularly exercised
individuals, and acromegaly patients.
Conditions with Increased [Insulin]:[GH] Ratio
Increased [insulin]:[GH] ratio is usually seen in conditions of adult GH deciency (AGHD), aging,
obese polycystic ovary syndrome (PCOS), and sleep disorders such as sleep deprivation and ob-
structive sleep apnea (OSA) (Table 1, lower panel). All show a profound reduction of GH [6265],
but with different changes in insulin secretion (elevated in AGHD [43], obese PCOS [66], OSA [67],
and in some aging individuals [68], whereas this is unaltered in sleep deprivation [69,70] and other
aging individuals [71]). Expectedly, aging is accompanied with reduced energy expenditure and
increased fat mass [72]. AGHD [73] and obese PCOS [74] patients have increased fat mass com-
pared with age-, sex-, height-, and weight-matched healthy adults, whereas both sleep depriva-
tion and OSA patients have a strong correlation with obesity [75] and show reduced ability to lose
weight following lifestyle intervention [7678]. In terms of energy expenditure, obese PCOS
patients have reduced postprandial thermogenesis [79]. Although studies show no signicant
changes in the energy expenditure of AGHD patients compared with body weight-matched
obese individuals [73,80], this is likely due to adaptation of energy intake and expenditure after
long-term GH deciency. At the onset of the GH deciency, patients may have reduced energy
expenditure, as shown in a mouse model of adult-onset, isolated GH deciency [81]. In sleep dis-
orders, one study shows that acute sleep deprivation reduces both resting and postprandial en-
ergy expenditure [82]; others show increased energy expenditure, which is likely to be a result of
more awake hours during the night and increased food intake [75].
Simple obesity is also a condition in which hyperinsulinemia is accompanied by low GH secretion
[83], as well as increased fat mass. However, it is difcult to determine energy expenditure accu-
rately because of the lack of proper controls. It would be important to look at the energy expen-
diture difference between obese and non-obese individuals with matched muscle mass because
muscle is the major organ that contributes to energy expenditure in humans. Nevertheless,
because of the high body weight and fat percentage in obese individuals, it is speculated that
these individuals may have reduced energy expenditure and increased fat accumulation.
Although insulinoma patients show increased insulin secretion, neither GH secretion nor energy
expenditure has been studied in this rare disease. Other diseases with reduced GH secretion
or action (e.g., childhood-onset GH deciency, Turner syndrome, and Laron syndrome) are not
discussed because the signicantly reduced height of these patients results in a lack of height-
and body weight-matched healthy controls.
Disease Therapies That Directly Modify Insulin or GH Levels
In addition to the changes in endogenous insulin/GH discussed above, alterations in the circulat-
ing levels of insulin/GH seen in diseased individuals following pharmacological or surgical inter-
vention also support application of the [insulin]:[GH] ratio to energy expenditure and fat
accumulation. Following long-term insulin or sulfonylurea treatment, the [insulin]:[GH] ratio of
T2D patients tends to be increased, followed by a reduction in resting metabolic rate [84]or
total energy expenditure [85]. The high basal energy expenditure of T1D patients returns to
normal following insulin administration [55], with a concurrent increase in the [insulin]:[GH] ratio.
Increased insulin levels as a result of either insulin or sulfonylurea treatment in T2D [86], as well
asinsulininjectioninT1D[87], lead to increased fat mass. In terms of changes in GH levels,
GH-decient patients show increased energy expenditure [80,88] and decreased fat accumula-
tion [88] after GH treatment compared with pretreatment. On the other hand, following reduction
of excessive GH secretion by surgical removal of a GH-releasing tumor, which is expected to
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 7
increase the [insulin]:[GH] ratio, acromegaly patients show reduced energy expenditure and basal
metabolic rate [50], and display a tendency to weight-gain [89].
Evidence from Mouse Models
Similar to the above mentioned studies in humans, rodent studies also support the association be-
tween the [insulin]:[GH] ratio and energy metabolism. Reduction of Ins1 expression in an Ins2 null
background leads to increased energy expenditure and resistance to diet-induced obesity when
fed with a high-fat diet [90]. Although GH levels were not measured in this model, they are expected
to increase because of attenuation of hyperinsulinemia, leading to a reduced [insulin]:[GH] ratio. The
[insulin]:[GH] ratio is increased in adult-onset GH deciency in mice, followed by reduced energy
expenditure and increased fat mass [81]. With bovine (b)GH overexpression, bGH mice have
N400-fold higher GH levels [91] and ~10-fold higher insulin levels [92] compared with wild-type lit-
termates, thus displaying a decreased [insulin]:[GH] ratio. Similarly to acromegaly patients, these
mice show both increased energy expenditure and decreased fat percentage [92]. A recent
study shows that sodium/glucose cotransporter 2 inhibitor (SGLT2i) treatment reduces
hyperinsulinemia and restores GH secretion in an obese mouse model through inhibition of glucose
reabsorption in the kidney [93], leading to a reduced [insulin]:[GH] ratio. The hormonal changes
contribute to improved insulin sensitivity, increased lipid usage, and reduced fat accumulation [93].
To summarize, various physiological and pathophysiological conditions support a negative
correlation between [insulin]:[GH] ratio and energy expenditure, and a positive correlation with
fat accumulation. Changes in this balance may lead to improvement or deterioration of the
metabolic conditions.
Potential Mechanisms of InsulinGH Balance in Regulating Energy Expenditure
Apart from the effects of different [insulin]:[GH] ratios in various diseases/treatments on energy
expenditure, some clinical studies show that blocking GH receptor signaling by pegvisomant
reduces basal energy expenditure in healthy individuals [94], acromegaly patients [95], and
insulin-resistant non-diabetic men [96]. These results suggest a direct effect of GH on energy
expenditure. The mechanisms behind the changes in energy expenditure as a function of the
[insulin]:[GH] ratio may be associated with alterations of mitochondrial function, BAT function,
and WAT browning by either insulin or GH.
Mitochondria are the primary site of energy production in the cell. The effects of insulin on mito-
chondria seem to depend on overall insulin sensitivity. Reduced mitochondria function is seen
in insulin-resistant T2D patients [97] and in mice with disruption of insulin receptor signaling
[98], whereas increased mitochondrial function is observed in healthy individuals with acute
insulin infusion [97]. Unlike insulin, acute GH administration increased mitochondrial function
and gene expression in both lean and obese humans [99,100]. Moreover, a GH-decient
model (spontaneous dwarf rat) shows decreased mitochondrial area in hepatocytes [101], and
GH supplementation increases mitochondria number in these isolated hepatocytes [102].
Although one study indicates that GH can accumulate in the mitochondria after acute GH administra-
tion, and inhibits mitochondrial function in isolated mitochondria [103], this study is limited to isolated
mitochondria, and may not reect the overall cell-based mechanisms. Therefore, the overall effect of
GH seems to increase mitochondrial function, but the exact mechanism remains unclear.
BAT and browning WAT are also essential contributors to energy expenditure. It has been shown
that JAK2 is crucial for the induction of UCP1 in BAT in response to cold exposure, high-fat diet
feeding, and adrenergic stimulation [104]. GH treatment increases UCP1, 2 and 3 expression in
BAT and WAT of obese KK-Ay mice [105]. In vitro study shows that GH treatment increases
Trends in Endocrinology & Metabolism
8Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
UCP1 expression in 3T3-L1 cells [106]. A recent animal study reveals that GH receptor signaling
is necessary for the induction of beige fat through STAT5 induction of ADRB3 [16]. In addition, GH
also supports beige fat by increasing both circulating and local adipose tissue production of
FGF21 [16]. However, some studies in mouse models with reduced GH secretion show enlarged
BAT or increased BAT function [107]. The reason is unknown, although it may relate to the smaller
size of these mice, hence increased cold stress. On the other hand, hyperinsulinemia reduces
BAT markers as well as mitochondrial content and activity in vitro and in vivo [108]. In high-fat-
fed mice, reducing hyperinsulinemia by reducing βcell insulin gene dosage leads to increased en-
ergy expenditure and UCP1 expression in WAT [90]. These results indicate that GH may increase
the thermogenic function of BAT and WAT browning, thus increasing energy expenditure,
whereas hyperinsulinemia has the opposite effect.
Treatment of Obesity by Modulating Insulin or GH
Acknowledging the effects of insulinGH balance on substrate and energy metabolism, it is likely
that modifying insulin or GH levels could be a useful therapeutic approach to treat obesity.
Although no drug is currently approved to treat obesity by directly targeting the insulin/GH
balance, there are several clinical studies with promising outcomes. We review here clinical
studies that directly reduce insulin (diazoxide) or increase GH [recombinant human GH (rhGH)
and tesamorelin], and summarize the pros and cons of these treatments (Table 2).
Table 2. Trials of Diazoxide and rhGH Therapy in Obesity
Individuals BMI Dose Duration Fasting
insulin
Oral glucose-
tolerance test
(OGTT) insulin
Fat
mass
Fasting
blood
glucose
(FBG)
Glucose
in OGTT
Insulin
resistance
Other Refs
Diazoxide
24 obese
adults
~40 2 mg/kg/day,
tid
a
(maximum
200 mg/day)
8 weeks ↓↓ ↔↔ With low-
calorie diet
[110]
35 obese
adults
~42 2 mg/kg/day,
tid (maximum
200 mg/day)
8 weeks ↔↓ ↔ ↑ Baseline
insulin does
not match
between
treatment and
placebo group
[112]
18 obese
men
34 50300 mg
tid
6 months ↓↓ ↑ ↑ ↓ With lifestyle
intervention
[111]
44 obese
men
~35 100350 mg
bid
a
6 months ↓↓ ↑ ↑ ↓ With lifestyle
intervention
[109]
rhGH
50 obese
women
35 1.7 ± 0.1
mg/day
(mean dose)
6 months ↓↑ ↑ [116]
62 obese
men
37 1.1 ± 0.08
mg/day
(nal dose)
6 months ↓↔ ↔ Improve
mitochondrial
function
[100]
15 obese
adults
38 0.49 ± 0.07
mg/day
(nal dose)
6 weeks ↓↔ ↔ Decrease
adipocyte size
[117]
50 obese
women
34 1.3 ± 0.5
mg/day
(mean dose)
3 months ↓↑[118]
a
Abbreviations: bid, twice per day; tid, three times per day.
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 9
Treatment of Obesity by Reducing Insulin
Diazoxide inhibits insulin secretion by opening ATP-sensitive potassium (K
ATP
) channels on the
pancreatic βcells. It may recover the suppressed GH secretion in obese individuals through
reducing hyperinsulinemia, thus lowering the [insulin]:[GH] ratio, although this needs to be
conrmed.
Clinical trials of diazoxide (Table 2) show mixed results in terms of glucose and lipid metabolism;
three studies showed reduced fat mass with or without slightly impaired glucose metabolism
[109111], and one showed no change in fat mass [112]. Importantly, no study has reported
any loss of lean mass during diazoxide treatment. The preservation of lean mass may be associated
with recovery of impaired GH secretion. Interestingly, treating obese children with hypothalamic/
pituitary lesions with diazoxide fails to achieve body weight reduction [113]. This could be due to
inability to recover GH, and thus strengthens the importance of GH in combating obesity. To
summarize, diazoxide treatment shows a signicant effect in reducing fat mass and improving
insulin sensitivity in most clinical trials, at the cost of slightly impaired glucose tolerance.
Treatment of Obesity by Increasing GH
The rhGH and tesamorelin treatments to increase GH levels in obesity have been thoroughly
reviewed by Berryman et al. [83] which includes clinical trials from 1967 to 2013. We summarize
the relevant parameters in additional studies after 2013 in Table 2. The overall conclusion is that
rhGH treatment reduces fat mass, increases lean mass, does not change body weight, and may
cause slightly impaired glucose metabolism in the short term.
However, although clinical trials show a promising benet of GH in treating obesity, there
remain many limitations relating to mimicking a physiological equivalent GH pattern. rhGH
injection induces a large plateau for ~48 h, which is different from the physiological pulsatile
secretion in which there are low levels of basal secretion and large secretory pulses at rhythmic
intervals [6]. This may result in different effects on substrate and energy metabolism, given that
pulsatile and continuous GH have different effects on GH receptor and STAT5 activation [7].
Tesamorelin is a GH-releasing hormone (GHRH) analog that has been used to treat HIV-
associated lipodystrophy. It increases GH secretion while preserving the pulsatile secretion
pattern of GH [114]. Clinical trials show that tesamorelin signicantly reduces fat mass in obese
individuals without compromising glucose metabolism [83,115]. Although this is encouraging,
the long-term effects on obesity and the cost will need to be considered.
To summarize, rhGH injection signicantly reduces fat mass at the cost of slightly impaired glucose
metabolism, whereas tesamorelin shows fat-reducing effects but has little inuence on glucose
metabolism. We encourage the further development of lower-cost drugs that can increase pulsatile
GH secretion. These may exert fat mass reduction without impairing glucose metabolism.
Concluding Remarks and Future Perspectives
To summarize, from an evolutionary standpoint, insulin promotes energy storage when food is
plentiful, whereas GH promotes lipid usage to protect proteinwhen food is sparse. In the contem-
porary world with excess food supply, the [insulin]:[GH] ratio increases and is related to an in-
crease in obesity. Given their synergetic effects on protein preservation, and their opposing
effects on glucose and lipid metabolism, as well as their reciprocal regulation of each others se-
cretion, insulin and GH should always be investigated concurrently to understand the complex
hormonal network in obesity. From analysis of the [insulin]:[GH] ratio in conditions of physiological,
pathophysiological, and pharmacological challenge, we conclude that the [insulin]:[GH] ratio is
negatively correlated with energy expenditure and is positively correlated with fat accumulation.
Outstanding Questions
What are the molecular mechanisms
by which the insulinGH balance regu-
lates energy expenditure?
Is hyperinsulinemia the dominant
factor that causes reduced GH secre-
tion in obesity?
To apply the ratio of insulin to GH in the
clinic, it is crucial to dene the way in
which insulin and GH levels are col-
lected. Although fasting insulin is
widely accepted for the basal state in-
sulin level, GH measurement is difcult
because of its pulsatile secretion pat-
tern. Is total IGF-1 a suitable surrogate
marker for the 'biologically effective'
GH?
What is the effect of different patterns
of GH (e.g., physiological pulsatile
pattern and 'plateau pattern' by rhGH
injection) on glucose, lipid, and energy
metabolism? Is there a way to supple-
ment GH in a specicpatternwhich
retains fat-reducing effect but mini-
mizes the negative effect on glucose
metabolism?
Trends in Endocrinology & Metabolism
10 Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
In addition, some clinical trials using pharmacological approaches to reduce insulin or increase
GH levels show benecial effects on obesity.
Therefore, the [insulin]:[GH] ratio could be a potential biomarker for predicting energy expenditure
and the development of obesity in clinical practice. Although fasting insulin is widely accepted as
an indicator of basal state insulin level, the choice of a suitable indicator for GH needs further
study. Random and stimulated GH are commonly used in the clinic. However, they have limited
physiological meaning because of the pulsatile secretion pattern of GH. Measuring the 24 h
secretion prole of GH is the gold standard, but is a complex sampling process and difcult
to use clinically. Therefore, it is crucial to nd a surrogate marker to show the 'biologically effective'
GH. Total IGF-1 may be a potential marker, together with N-terminal pro-peptide of type
3 collagen, as used by World Anti-Doping Agency (WADA) for human GH testing in sport.
Whether this is strongly correlated with pulsatile or total or basal GH secretion needs to
be tested in individuals without acromegaly or GH deciency.
There are still many unknowns in the mechanism of the regulation and effects of insulin and GH,
including the mechanisms underlying reduced GH secretion in obesity and the different effects of
physiological (pulsatile) or therapeutic rhGH injection on substrate and energy metabolism.
Further basic studies will be necessary to answer these questions (see Outstanding Questions).
Acknowledgments
Z.H. is the recipient of a China Scholarship Council PhD scholarship and a University of Queensland international PhD scholarship.
This work is supported by Grants to C.C. from NHMRC and University of Queensland.
References
1. Clasey, J.L. et al. (2001) Abdominal visceral fat and fasting
insulin are important predictors of 24-hour GH release inde-
pendent of age, gender, and other physiological factors.
J. Clin. Endocrinol. Metab. 86, 38453852
2. Moller, N. and Jorgensen, J.O. (2009) Effects of growth
hormone on glucose, lipid, and protein metabolism in human
subjects. Endocr. Rev. 30, 152177
3. Cornford, A.S. et al. (2011) Rapid suppression of growth hor-
mone concentration by overeating: potential mediation by
hyperinsulinemia. J. Clin. Endocrinol. Metab. 96, 824830
4. Tan, H.Y. et al. (2016) Hyperphagia in male melanocortin 4
receptor decient mice promotes growth independently of
growth hormone. J. Physiol. 594, 73097326
5. Pørksen, N. et al. (2002) Pulsatile insulin secretion: detection,
regulation, and role in diabetes. Diabetes 51, S245S254
6. Winer, L.M. et al. (1990) Basal plasma growth hormone levels
in man: new evidence for rhythmicity of gr owth hormone
secretion. J. Clin. Endocrinol. Metab. 70, 16781686
7. Huang, L. et al. (2019) Rhythmic growth hormone secretion in
physiological and pathological conditions: lessons from rodent
studies. Mol. Cell. Endocrinol. 498, 110575
8. Siddle, K. (2011) Signalling by insulin and IGF receptors:
supporting acts and new players. J. Mol. Endocrinol. 47,
R1R10
9. Brooks, A.J. and Waters, M.J. (2010) The growth hormone
receptor: mechanism of activation and clinical implications.
Nat. Rev. Endocrinol. 6, 515525
10. Woele, J. et al. (2003) Acute control of insulin-like growth
factor-I gene transcription by growth hormone th rough
Stat5b. J. Biol. Chem. 278, 2269672202
11. Argetsinger, L.S. et al. (1996) Growth hormone, interferon-gamma,
and leukemia inhibitory factor utilize insulin receptor substrate-2 in
intracellular signaling. J. Biol. Chem. 271, 2941529421
12. Ridderstrale, M. et al. (1995) Growth horm one stimulates the
tyrosine phosphorylation of the insulin receptor substrate-1
and its association with phosphatidylinositol 3-kinase in
primary adipocytes. J. Biol. Chem. 270, 34713474
13. Yamauchi, T. et al. (1998) Growth hormone and prolactin stim-
ulate tyrosine phosphorylation of insulin receptor substrate-1,
-2, and -3, their associ ation with p85 phosph atidylinositol 3-
kinase (PI3-kinase), and concomitantly PI3-kinase activation
via JAK2 kinase. J. Biol. Chem. 273, 1571915726
14. Haeusler, R.A. et al. (2017) Biochemical and cellular properties
of insulin receptor signalling. Nat. Rev. Mol. Cell Biol. 19, 31
15. Yang, S. et al. (2004) Effects of growth hormone on the func-
tion of β-adrenoceptor subtypes in rat adipocytes. Obes.
Res. 12, 330339
16. Nelson, C.N. et al. (2018) Growth hormone activated STAT5 is
required for induction of beige fat in vivo. Growth Hormon. IGF
Res. 42, 4051
17. Stralfors, P. et al. (1984) Hormonal regulation of hormone-
sensitive lipase in intact adipocytes: identication of phosphor-
ylated sites and effects on the phosphorylation by lipolytic
hormones and insulin. Proc.Natl.Acad.Sci.U.S.A.81,
33173321
18. Sharma, V.M. et al. (2019) Growth hormone acts along the
PPARgammaFSP27 axis to stimulate lipolysis in human
adipocytes. Am. J. Physiol. Endocrinol. Metab. 316,
E34E42
19. Sears, B. and Perry, M. (2015) The role of fatty acids in insulin
resistance. Lipids Health Dis. 14, 121
20. LeRoith, D. and Yakar, S. (2007) Mechanisms of disease: met-
abolic effects of growth hormone and insulin-like growth factor
1. Nat. Clin. Pract. Endocrinol. Metab. 3, 302310
21. Liu, Z. et al. (2016) G rowth hormon e control o f hepatic lipi d
metabolism. Diabetes 65, 35983609
22. Sos, B.C. et al. (2011) Abrogation of growth hormone secre-
tion rescues fatty liver in mice with hepatocyte-specic deletion
of JAK2. J. Clin. Invest. 121, 14121423
23. Barclay, J.L. et al. (2011) GH-dependent STAT5 signaling plays
an important role in hepatic lipid metabolism. Endocrinolog y
152, 181192
24. Chhabra, Y. et al. (2019) Lo ss of growth hormone-m ediated
signal transducer and activator of transcription 5 (STAT5)
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 11
signaling in mice results in insulin sensitivity with obesity.
FASEB J. 33, 64126430
25. Kim, Y.D. et al. (2012) Orphan nuclear receptor small heterodi-
mer partner negatively regulates growth hormone-mediated
induction of hepatic gluconeogenesis through inhibition of
signal transducer and activator of transcription 5 (STAT5)
transactivation. J. Biol. Chem. 287, 3709837108
26. Kim, Y.D. et al. (2012) Metformin inhibits growth hormone-
mediated hepatic PDK4 gene expression through induction
of orphan nuclear receptor small heterodimerpartner. Diabetes
61, 24842494
27. Hoybye, C. et al. (2008) Contribution of gluconeogenesis and
glycogenoly sis to hepati c glucose prod uction in acro megaly
before and after pituitary microsurgery. Horm. Metab. Res.
40, 498501
28. Saxton,R.A.andSabatini,D.M.(2017)mTORsignalingin
growth, metabolism, and disease. Cell 168, 960976
29. Hayashi, A.A. and Proud, C.G. (2007) The rapid activation of
protein synthesis by growth hormone requires signaling
through mTOR. Am.J.Physiol.Endocrinol.Metab.292,
E1647E1655
30. Kim, H. et al. (2005) Intact insulin and insulin-like growth factor-
I receptor signaling is required for growth hormone effects on
skeletal muscle growth and function in vivo.Endocrinology
146, 17721779
31. Norrelund, H. et al. (2003) The decisive role of free fatty acids
for protein conservation during fasting in humans with and
without growth hormone. J. Clin. Endocrinol. Metab. 88,
43714378
32. Dehkhoda, F. et al. (2018) The growth hormone receptor:
mechanism of receptor activation, ce ll signaling, and physio-
logical aspects. Front. Endocrinol. 9, 35
33. Ji, S. et al. (1999) Insulin inhibits growth hormone signaling via
the growth hormone receptor/JAK2/STAT5B pathway. J. Biol.
Chem. 274, 1343413442
34. Xu, J. and Messina, J.L. (2009) Crosstalk between growth
hormone and insulin signaling. Vitam. Horm. 80, 125153
35. Barbour, L.A. et al. (2005) Increased P85alpha is a potent neg-
ative regulator of skeletal muscle insulin signaling and induces
in vivo insulin resistance associated with growth hormone
excess. J. Biol. Chem. 280, 3748937494
36. Gao, Y. et al. (2013) The role of PTEN in chronic growth
hormone-induced hepatic insulin resistance. PLoS One 8,
e68105
37. Ueki, K. et al. (2004 ) Central role o f suppressors of cytokine
signaling proteins in hepatic steatosis, insul in resistance, and
the metabolic syndrome in the mouse. Proc. Natl. Acad. Sci.
U. S. A. 101, 1042210427
38. Leung, K.-C. et al. (2000) Insulin regulation of human he patic
growth hormone receptors: divergent effects on biosyn thesis
and surface translocation. J. Clin. Endocrinol. Metab. 85,
47124720
39. Hoeck, H.C. et al. (2000) Dia gnosis of growth hormo ne (GH)
deciency in adults with hypothalamic-pituitarydisorders: com-
parison of test results using pyridostigmine plus GH-releasing
hormone (GHRH), cloni dine plus GHRH, and in sulin-induced
hypoglycemia as GH secretagogues. J. Clin. Endocrinol.
Metab. 85, 14671472
40. Gahete, M.D. et al. (2013) Insulin and IGF-I inhibit GH synthesis
and release in vitro and in vivo by separate mechanisms.
Endocrinology 154, 24102420
41. Powell, D.R. et al. (1991) Insulin inhibits transcription of the
human gene for insulin-like growth factor-binding protein-1.
J. Biol. Chem. 266, 1886818876
42. Husbands, S. et al. (2001) Increased insulin sensitivity in
young, growth hormone decient children. Clin. Endocrinol.
55, 8792
43. Johansson, J.O. et al. (1995) Growth hormone-decient adults
are insulin-resistant. Metabolism 44, 11261129
44. Wu, Y. et al. (2011) Growth hormone receptor regulates βcell
hyperplasia and glucose-stimulated insulin secretion in obese
mice. J. Clin. Invest. 121, 24222426
45. Oliveira, C.R. et al. (2012) Insulin sensitivity and beta-cell func-
tion in adults with lifetime, untreated isolated growth hormone
deciency. J. Clin. Endocrinol. Metab. 97, 10131019
46. Holly, J.M. et al. (1988) The role of growth hormone in diabetes
mellitus. J. Endocrinol. 118, 353364
47. FRYSTYK, J. (2010) Exercise and the growth hormoneinsulin-
like growth factor axis. Med. Sci. Sports Exerc. 42, 5866
48. Bird, S.R. and Hawley, J.A . (2017) Upda te on the effec ts of
physical activity on insulin sensitivity in human s. BMJ Open
Sport Exerc. Med. 2, e000143
49. Foss, M.C. et al. (1991) Peripheral glucose metabolism in
acromegaly. J. Clin. Endocrinol. Metab. 72, 10481053
50. Guo, X. et al. (2018) Pre- and postoperative body composition
and metabolic characteristics in patients with acromegaly: a
prospective study. Int. J. Endocrinol. 2018, 4125013
51. Gómez, J.M. et al. (2001) Body composition assessment in
type 1 diabetes mellitus patients over 15 years old. Horm.
Metab. Res. 33, 670673
52. Rosenfalck, A.M. et al. (2002) Body composition in adults with
type 1 diabetes at onset and during the rst year of insulin
therapy. Diabet. Med. 19, 417423
53. Godfrey, R.J. et al. (2003) The exercise-induced growth
hormone response in athletes. Sports Med. 33, 599613
54. O'Sullivan, A.J. et al. (1994) Body composition and energy
expenditure in acromegaly. J. Clin. Endocrinol. Metab. 78,
381386
55. Nair, K.S. et al. (1984) Increased energy expenditure in poorly
controlled type 1 (insulin-dependent) diabetic patients.
Diabetologia 27, 1316
56. Karakelides, H. et al. (2007) Effect of insulin deprivation on
muscle mitochondrial ATP production and gene transcript
levels in type 1 diabetic subjects. Diabetes 56, 26832689
57. Melby, C. et al. (1993) Effect of acut e resistance exercis e on
postexercise e nergy expenditur e and resting meta bolic rate.
J. Appl. Physiol. 75, 18471853
58. Dolezal, B.A. and Potteiger, J.A. (1998) Concurrent resistance
and endurance training inuence basal metabolic rate in
nondieting individuals. J. Appl. Physiol. 85, 695700
59. Tremblay, A. et al. (1986) The effect of exercise-training
on resting metabolic rate in lean and moderately obese
individuals. Int. J. Obes. 10, 511517
60. Moller, N. et al. (1992) Basal- and insulin-stimulated substrate
metabolism in patients with active acromegaly before and
after adenomectomy. J. Clin. Endocrinol. Metab. 74,
10121019
61. O'Sullivan, A.J. et al. (1995) Energy metabolism and substrate
oxidation in acromegaly. J. Clin. Endocrinol. Metab. 80,
486491
62. Steyn, F.J. et al. (2016) Neuroendocr ine regulation of growth
hormone secretion. Compr. Physiol. 6, 687735
63. Morales, A.J. (1997) Role of growth hormone in polycystic
ovarian syndrome. Semin. Reprod. Endocrinol. 15, 177182
64. Ritsche, K. et al. (2014) Exercise-induced growth hormone
during acute sleep deprivation. Physiol. Rep. 2, e12166
65. Lanfranco, F. et al. (2010) Growth hormone/insulin-like growth
factor-I axis in obstructive sleep apnea syndrome: an update.
J. Endocrinol. Investig. 33, 192196
66. Dunaif, A. (1997) Insulin resistance and the polycystic ovary
syndrome: mechanism and implications for pathogenesis.
Endocr. Rev. 18, 774800
67. Ip, M.M. et al. (2002) Obstructive sleep apnea is independently
associated with insulin resistan ce. Am.J.Respir.Crit.Care
Med. 165, 670676
68. Fink, R.I. et al. (1983) Mechanisms of insulin resistance in
aging. J. Clin. Invest. 71, 15231535
69. Corssmit, E.P.M. et al. (2010) A single night of partial sleep
deprivation induces insulin resistance in multiple metabolic
pathways in healthy subjects. J. Clin. Endocrinol. Metab. 95,
29632968
70. Buxton, O.M. et al. (2010) Sleep restriction for 1 week re-
duces insulin sensitivity in healthy men. Diabetes 59,
21262133
71. Chang, A.M. and Halter, J.B. (2003) Aging and insulin
secretion. Am. J. Physiol. Endocrinol. Metab. 284, E712
72. St-Onge, M.- P. and Gallag her, D. (2010) Bod y compositi on
changes wit h aging: the c ause or the re sult of alter ations in
metabolic rate and macronutrient oxidation? Nutrition
(Burbank) 26, 152155
Trends in Endocrinology & Metabolism
12 Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx
73. Hoffman, D.M. et al. (1995) Adults with growth hormone
deciency have abnormal body composition but normal
energy metabolism. J. Clin. Endocrinol. Metab. 80, 7277
74. Kirchengast, S. and Huber, J. (2001) Body composition char-
acteristics and body fat distributio n in lean women with poly-
cystic ovary syndrome. Hum. Reprod. 16, 12551260
75. St-Onge, M.-P. and Shecht er, A. (2014) Sleep disturban ces,
body fat distributi on, food intake and /or energy expendi ture:
pathophysiological aspects. Horm. Mol. Biol. Clin. Inv est. 17,
2937
76. Nedeltcheva, A. V. et al. (2010) Insufc ient sleep underm ines
dietary efforts to reduce adiposity. Ann. Intern. Med. 153,
435441
77. Chaput, J.P. and Tremblay, A. (2012) Sleeping habits predict
the magnitude of fat loss in adults exposed to moderate caloric
restriction. Obes. Facts 5, 561566
78. Thomson, C.A. et al. (2012) Relationship between sleep quality
and quantity and weight loss in women participating in a
weight-loss intervention trial. Obesity (Silver Spring) 20,
14191425
79. Robinson, S. et al. (1992) Postprandial thermogenesis is
reduced in polycystic ovary syndrome and is associated with
increased insulin resistance. Clin. Endocrinol. 36, 537543
80. Chong, P.K. et al. (1994) Energy expenditure and body com-
position in growth hormone decient adult s on exogenous
growth hormone. Clin. Endocrinol. 40, 103110
81. Luque, R.M. et al. (2011) Metabolic impact of adult-onset, iso-
lated, growth hormone deciency (AOiGHD) due to destruction
of pituitary somatotropes. PLoS One 6, e15767
82. Benedict, C. et al. (2011) Acute sleep deprivation reduces
energy expenditure in healthy men. Am.J.Clin.Nutr.93,
12291236
83. Berryman, D.E. et al. (2013) The GH/IGF-1 axis in obesity:
pathophysiology and therapeutic considerations. Nat. Rev.
Endocrinol. 9, 346356
84. Chong, P.K. et al. (1995) Energy expenditure in type 2 diabetic
patients on metformin and sulphonylurea therapy. Diabet.
Med. 12, 401408
85. Nakaya, Y. et al. (1998) Respirator y quotient in patient s with
non-insulin-dependent diabetes mellitus tre ated with insulin
and oral hypoglycemic agents. Ann. Nutr. Metab. 42, 333340
86. Barnett, A. et al. (2007) A revi ew of the effects of
antihyperglycaemic age nts on body weight: the po tential of
incretin targetedtherapies. Curr.Med.Res.Opin.23, 14931507
87. Mottalib, A. et al. (2017) Weight management in patients with
type 1 diabetes and obesity. Curr. Diabetes Rep. 17, 92
88. Straetemans, S. et al. (2019) Effect of growth hormone treat-
ment on energy expenditure and its relation to rst-year growth
response in children. Eur. J. Appl. Physiol. 119, 409418
89. Reyes-Vidal, C. et al. (2014) Prospective study of surgical treat-
ment of acromegaly: effects on ghrelin, weight, adiposity, and
markers of CV risk. J. Clin. Endocrinol. Metab. 99, 41244132
90. Mehran, A.E. et al. (2012) Hyperinsulinemia drives diet-induced
obesity independently of brain insulin production. Cell Metab.
16, 723737
91. Palmer, A.J.et al. (2009) Age-relatedchanges in body composi-
tion of bovine growth hormone transgenic mice. Endocrinology
150, 13531360
92. Olsson, B. et al. (2005) Bovine growth hormone transgenic
mice are resistant to diet-induced obesity but develop
hyperphagia, dyslipidemia, and diabetes on a high-fat diet.
Endocrinology 146, 920930
93. Huang, Z. et al. (2020) Dapagliozin restores insulin and
growth hormone secretion in obese mice. J. Endocrinol.
245, 112
94. Moller, L. et al. (2009) Impact of growth hormone recep-
tor blockade on substrate metabolism during fasting i n
healthy subjects. J. Clin. Endocrinol. Metab. 94,
45244532
95. Lindberg-Larsen, R. et al . (2007) The impact of pegvisoman t
treatment on substrate metabolism and insulin sensitivity in
patients with acromegaly. J. Clin. Endocrinol. Metab. 92,
17241728
96. Lee, A.P. et al. (2017) Growth hormone receptor antagoni sm
with pegvisomant in insulin resistant non-diabetic men: a
phase II pilot study. F1000Res. 6, 614
97. Asmann, Y.W. et al. (2006) Skeletal muscle mitochondrial functions,
mitocho ndrial D NA copy nu mbers, a nd gene tr anscrip t proles in
type 2 diabetic and nondiabetic subjects at equal levels of low or
high insulin and euglycemia. Diabetes 55, 33093319
98. Cheng, Z. et al. (2009) Foxo1 integrates insulin signaling with
mitochondrial function in the liver. Nat. Med. 15, 13071311
99. Makimura, H. et al. (2011) The association of growth hormone
parameters with skeletal muscle phosphocreatine recovery in
adult men. J. Clin. Endocrinol. Metab. 96, 817823
100. Bredella, M.A. et al. (2013) Effects of GH on body composition
and cardiovascular risk markers in young men with abdominal
obesity. J. Clin. Endocrinol. Metab. 98, 38643872
101. Nishizawa, H. et al. (2012) GH-independent IGF-I action is
essential to prevent the development of nonalcoholic
steatohepatitis in a GH-decient rat model. Biochem. Biophys.
Res. Commun. 423, 295300
102. Ishikawa, M. et al. (2010) Novel functional hepatocyte cell line
derived from spontaneous dwarf rat: model of growth hormone
function in vitro. Hum. Cell 23, 164172
103. Ardail, D. et al. (2010) Growth hormone internalization in mitochon-
dria decreases respiratory chain activity. Neuroendocrinology 91,
1626
104. Dodington, D.W. et al. (2018) JAK/STAT emerging players in
metabolism. Trends Endocrinol. Metab. 29, 5565
105. Hioki, C. et al. (2004) Effects of growth hormone (GH) on mRNA
levels of uncoupling proteins 1, 2, and 3 in brown and white ad-
ipose tissues and skeletal muscle in obese mice. Horm. Metab.
Res. 36, 607613
106. Hayashi, M. et al. (2017) Effects of growth hormone on
uncoupling protein 1 in white adipos e tissues in obese mice.
Growth Hormon. IGF Res. 37, 3139
107. Darcy, J. et al. (2016) Brown adipose tissue function is
enhanced in long-lived, male ames dwarf mice. Endocrinology
157, 47444753
108. Rajan, S. et al. (2016) Chronic hyperinsulinemia reduces insulin
sensitivity and metabolic functions of brown adipocyte.
J. Endocrinol. 230, 275290
109. Loves, S. et al. (2018) Effects of diazoxide-mediated insulin
suppression o n glucose and li pid metabolis m in nondiabet ic
obese men. J. Clin. Endocrinol. Metab. 103, 23462353
110. Alemzadeh, R. et al. (1998) Benecial effect of diazoxide in
obese hyperinsulinemic adults. J. Clin. Endocrinol. Metab. 83,
19111915
111. van Boekel, G. et al. (2008) Weight loss in obese men by
caloric restriction and high-dose diazoxide-mediated insulin
suppression. Diabetes Obes. Metab. 10, 11951203
112. Due, A. et al. (2007) No effect of inhibition of insulin secretion by
diazoxide on weight loss in hyperinsulin aemic obese subjects
during an 8-week weight-loss diet. Diabetes. Obes. Metab. 9,
566574
113. Brauner, R. et al. (2016) Diazoxide in children with obesity
after hypothalamic-pituitary lesions: a randomized, placebo-
controlled trial. J. Clin. Endocrinol. Metab. 101, 48254833
114. Stanley, T.L. et al. (2011) Effects of a growth hormone-releasing
hormone analog on endogenous GH pulsatility and insulin sensi-
tivity in healthy men. J. Clin. Endocrinol. Metab. 96, 150158
115. Falutz, J. et al. (2010) Effects of tesamorelin (TH9507), a growth
hormone-releasing factor analog, in human immunodeciency
virus-infected patients with excess abdominal fat: a pooled
analysis of two multicenter, double-blind placebo-controlled
phase 3 trials wi th safety extens ion data. J. Cli n. Endocrinol.
Metab. 95, 42914304
116. Lin, E. et al. (2013) Effects of growth hormone withdrawal in
obese premenopausal women. Clin. Endocrinol. 78, 914919
117. Bredella, M.A. et al. (2017) GH administration decreases sub-
cutaneous abdominal adipocyte size in men with abdominal
obesity. Growth Hormon. IGF Res. 35, 1720
118. Dichtel, L.E. et al. (2018) The effect of growth hor mone on
bioactive IGF in overweight/obes e women. Growth H ormon.
IGF Res. 40, 2027
Trends in Endocrinology & Metabolism
Trends in Endocrinology & Metabolism, Month 2020, Vol. xx, No. xx 13
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr‐391−/−) as for GHR‐null (GHR−/−) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor β and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)‐1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin‐2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up‐regulation of IL‐15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.—Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone‐mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity. FASEB J. 33, 6412–6430 (2019). www.fasebj.org
Article
Full-text available
Purpose The effects of growth hormone (GH) treatment on linear growth and body composition have been studied extensively. Little is known about the GH effect on energy expenditure (EE). The aim of this study was to investigate the effects of GH treatment on EE in children, and to study whether the changes in EE can predict the height gain after 1 year. Methods Total EE (TEE), basal metabolic rate (BMR), and physical activity level (PAL) measurements before and after 6 weeks of GH treatment were performed in 18 prepubertal children (5 girls, 13 boys) born small for gestational age (n = 14) or with growth hormone deficiency (n = 4) who were eligible for GH treatment. TEE was measured with the doubly labelled water method, BMR was measured with an open-circuit ventilated hood system, PAL was assessed using an accelerometer for movement registration and calculated (PAL = TEE/BMR), activity related EE (AEE) was calculated [AEE = (0.9 × TEE) − BMR]. Height measurements at start and after 1 year of GH treatment were analysed. This is a 1-year longitudinal intervention study, without a control group for comparison. Results BMR and TEE increased significantly (resp. 5% and 7%). Physical activity (counts/day), PAL, and AEE did not change. 11 out of 13 patients (85%) with an increased TEE after 6 weeks of GH treatment had a good first-year growth response (∆height SDS > 0.5). Conclusions GH treatment showed a positive effect on EE in prepubertal children after 6 weeks. No effect on physical activity was observed. The increase in TEE appeared to be valuable for the prediction of good first-year growth responders to GH treatment.
Article
Full-text available
The lipolytic effects of GH have been known for half a century and play an important physiological role for substrate metabolism during fasting. In addition, sustained GH-induced lipolysis is causally linked to insulin resistance. However, the underlying molecular mechanisms remain elusive. In the present study, we obtained experimental data in human subjects and used human adipose-derived stromal vascular cells (hADSCs) as a model system to elucidate GH-triggered molecular signaling that stimulates adipose tissue lipolysis and insulin resistance in human adipocytes. We discovered that GH downregulates the expression of fat specific protein (FSP27), a negative regulator of lipolysis, by impairing the transcriptional ability of the master transcriptional regulator, peroxisome proliferator-activated receptor gamma (PPARγ) via MEK/ERK activation. Ultimately, GH treatment promotes phosphorylation of PPARγ at Ser273 and causes its translocation from nucleus to the cytosol. Surprisingly, FSP27 overexpression inhibited PPARγ Ser273 phosphorylation and promoted its nuclear retention. GH antagonist treatment had similar effects. Our study identifies a novel signaling mechanism by which GH transcriptionally induce lipolysis via MEK/ERK pathway that acts along PPARγ-FSP27 in human adipose tissue.
Article
The well-documented hormonal disturbance in a general obese population is characterised by an increase in insulin secretion and a decrease in growth hormone (GH) secretion. Such hormonal disturbance promotes an increase in fat mass, which deteriorates obesity and accelerates the development of insulin resistance and type 2 diabetes. While the pathological consequence is alarming, the pharmaceutical approach attempting to correct such hormonal disturbance remains limited. By applying an emerging anti-diabetic drug, the sodium-glucose cotransporter 2 inhibitor, dapagliflozin (1mg/kg/day for 10 weeks), to a hyperphagic obese mouse model, we observed a significant improvement in insulin and GH secretion as early as 4 weeks after the initiation of the treatment. Restoration of pathological disturbance of insulin and GH secretion reduced fat accumulation and preserved lean body mass in the obese animal model. Such phenotypic improvement followed with concurrent improvements in glucose and lipid metabolism, insulin sensitivity, as well as the expression of metabolic genes that were regulated by insulin and GH. In conclusion, 10 weeks of treatment with dapagliflozin effectively reduces hyperinsulinemia and restores pulsatile GH secretion in the hyperphagic obese mice with considerable improvement in lipid and glucose metabolism. Promising outcomes from this study may provide insights into drug intervention to correct hormonal disturbance in obesity to delay the diabetes progression.
Article
Evolutionally conserved in all mammalians, the release of GH occurs in a rhythmic pattern, characterized by several dominant surges (pulsatile GH) with tonic low inter-pulse levels (tonic GH). Such pulsatile secretion pattern is essential for many physiological actions of GH on different tissues with defined gender dimorphism. Rhythmic release of pulsatile GH is tightly controlled by hypothalamic neurons as well as peripheral metabolic factors. Changes of GH pattern occur within a range of sophisticated physiological and pathological settings and significantly contribute to growth, ageing, survival and disease predispositions. Precise analysis of GH secretion pattern is vitally important for a comprehensive understanding of the function of GH and the components that regulate GH secretion pattern.
Article
Objective The anti-obesity actions of growth hormone (GH) led us to investigate if GH signaling is able to regulate beige/brite fat development of white adipose tissue (WAT). Methods We studied WAT in GHR-391 mice engineered to be unable to activate STAT5 in response to GH, in mice with adipose specific deletion of GHR, in GHR−/− mice and in bGH transgenic mice. QPCR, immunoblots and immunohistochemistry were used to characterize WAT. The in vivo effects of β-3 adrenergic activation with CL-316,243 and that of FGF21 infusion were also studied. Results GHR-391 mice had lower surface temperature than WT, with deficiency in β-oxidation and beiging transcripts including Ucp1. Oxidative phosphorylation complex subunit proteins were decreased dramatically in GHR-391 inguinal white adipose tissue (iWAT), but increased in bGH iWAT, as were proteins for beige/brown markers. In accord with its lack of β-3 adrenergic receptors, iWAT of GHR-391 mice did not beige in response to administration of the β-3 specific agonist CL-316,243 in contrast to WT mice. GHR-391 mice are deficient in FGF21, but unlike WT, infusion of the purified protein was without effect on extent of beiging. Finally, fat-specific deletion of the GHR replicated the loss of beiging associated transcripts. Conclusion In addition to promoting lipolysis, our study suggests that GH is able to promote formation of beige adipose tissue through activation of STAT5 and induction of Adrb3. This sensitizes WAT to adrenergic input, and may contribute to the anti-obesity actions of GH.
Article
Introduction It has been suggested that stimulation of lipolysis by Diazoxide (DZX)-mediated insulin suppression may be useful as a treatment of obesity. However, the optimal dose to promote lipolysis without causing hyperglycemia is currently unknown. Aim To assess the effects of DZX in non-diabetic obese men on lipid and glucose metabolism. Methods Double-blind, placebo-controlled, 6-month trial in men with a BMI of 30 - 37.5 kg/m² treated with a combination of caloric restriction, a standardized exercise program, and DZX or placebo dose escalation. Results The mean maximal tolerated dose (MTD) of DZX was 422 ± 44 mg/day (range 200 – 700 mg/day). Dose limiting events were edema (n = 11), hyperglycemia (n = 6) and nausea (n = 2). After dose reduction to a level free of clinical side effects, DZX treatment was associated with a markedly greater decrease in fasting insulin levels than placebo (-72.3 ± 3.5 % vs -23.0 ± 12.6 %, P <0.001), and a significant improvement of blood pressure and plasma lipid levels. The decline in insulin levels occurred at the cost of a small rise in plasma glucose (0.6 ± 0.2 mmol/l vs -0.1 ± 0.1 mmol/l, P = 0.04) and HbA1c (0.2 ± 0.1 % vs 0.0 ± 0.1 %, P = 0.17). Conclusion In non-diabetic obese men, insulin levels can be reduced up to 70% without major metabolic side effects. The marked inter-subject variation in MTD indicates that DZX dose titration needs to be individualized.
Article
Objective Overweight/obesity is characterized by decreased growth hormone (GH) secretion whereas circulating IGF-I levels are less severely reduced. Yet, the activity of the circulating IGF-system appears to be normal in overweight/obese subjects, as estimated by the ability of serum to activate the IGF-I receptor in vitro (bioactive IGF). We hypothesized that preservation of bioactive IGF in overweight/obese women is regulated by an insulin-mediated suppression of IGF-binding protein-1 (IGFBP-1) and IGFBP-2, and by suppression of IGFBP-3, mediated by low GH. We additionally hypothesized that increases in bioactive IGF would drive changes in body composition with low-dose GH administration. Design Cross-sectional analysis and 3-month interim analysis of a 6-month randomized, placebo-controlled study of GH administration in 50 overweight/obese women without diabetes mellitus. Bioactive IGF (kinase receptor activation assay) and body composition (DXA) were measured. Results Prior to treatment, IGFBP-3 (r = −0.33, p = 0.02), but neither IGFBP-1 nor IGFBP-2, associated inversely with bioactive IGF. In multivariate analysis, lower IGFBP-3 correlated with lower peak stimulated GH (r = 0.45, p = 0.05) and higher insulin sensitivity (r = −0.74, p = 0.003). GH administration resulted in an increase in mean serum IGF-I concentrations (144 ± 56 to 269 ± 66 μg/L, p < 0.0001) and bioactive IGF (1.29 ± 0.39 to 2.60 ± 1.12 μg/L, p < 0.0001). The treatment-related increase in bioactive IGF, but not total IGF-I concentration, predicted an increase in lean mass (r = 0.31, p = 0.03) and decrease in total adipose tissue/BMI (r = −0.43, p = 0.003). Conclusions Our data suggest that in overweight/obesity, insulin sensitivity and GH have opposing effects on IGF bioactivity through effects on IGFBP-3. Furthermore, increases in bioactive IGF, rather than IGF-I concentration, predicted GH administration-related body composition changes. Clinical Trial Registration Number: NCT00131378.