Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Mol Cell Endocrinol. 2020 Sep 20;518:111038. doi: 10.1016/j.mce.2020.111038

Effect of Growth Hormone on Insulin Signaling

Rita Sharma 1, John J Kopchick 1,2,3, Vishwajeet Puri 1,3, Vishva M Sharma 1,3,ψ
PMCID: PMC7606590  NIHMSID: NIHMS1633712  PMID: 32966863

Abstract

Growth hormone (GH) is a pleiotropic hormone that coordinates an array of physiological processes, including effects on bone, muscle, and fat, ultimately resulting in growth. Metabolically, GH promotes anabolic action in most tissues except adipose, where its catabolic action causes the breakdown of stored triglycerides into free fatty acids (FFA). GH antagonizes insulin action via various molecular pathways. Chronic GH secretion suppresses the anti-lipolytic action of insulin and increases FFA flux into the systemic circulation; thus, promoting lipotoxicity, which causes pathophysiological problems, including insulin resistance. In this review, we will provide an update on GH-stimulated adipose lipolysis and its consequences on insulin signaling in liver, skeletal muscle, and adipose tissue. Furthermore, we will discuss the mechanisms that contribute to the diabetogenic action of GH.

Keywords: Insulin resistance, FSP27, CIDEC, Fat metabolism, lipolysis, Type 2 diabetes

Introduction

GH research and its use in clinics to cure growth disorders span nearly a century. Elegant work by Evans and Long in 1922 demonstrated that intraperitoneal injection of a fresh bovine anterior hypophyseal extract accelerated growth in rats (1). Later, studies by Houssay and colleagues in 1930s and early 1940s established that hypophyseal extracts or crude anterior pituitary extracts accelerated growth but also promoted diabetes in dogs and cats (25). In 1942, Houssay demonstrated that these pituitary extracts induced histological changes in many tissues and damaged pancreatic Langerhans islets, indicating their diabetogenic potential (6). After successful isolation of GH from the anterior pituitary by Li and colleagues in 1944 (7), a series of studies by multiple groups reported that GH has the potential to induce growth, diabetes, and hyperglycemia in animals (711). Rabinowitz et al in 1965 showed in humans that GH promotes diabetogenic action by antagonizing insulin action in muscle and adipose via promoting the release of free fatty acids (FFAs) from the adipose tissue (12). In 1963, human growth hormone (HGH) isolated from pituitary glands of cadavers was approved for clinical use to treat children who were unusually short due to pituitary disorders resulting in GH deficiency (D). In 1985, the U.S. Food and Drug Administration (FDA) approved recombinant (r) HGH to be used for clinical and research purposes (13).

The pleiotropic actions of GH involve multiple organs and physiological systems. GH exerts many metabolic effects that persist throughout life (14, 15). Kastin et al. showed that lipid mobilizing substances present in the hypothalamus and pituitary of mammals are involved in the central neuronal control of obesity and hypothesized that GH-releasing hormone (GHRH) might influence lipolysis secondarily by regulating pituitary hormones (e.g., GH) release (16). Growth promoting effects of GH on cartilage and bone are most notable, especially during the adolescent years (17). In addition to promoting growth, GH exerts profound anabolic actions during postnatal skeletal development, in-part through stimulating insulin-like growth factor-1(IGF-1) (17). GH induces skeletal IGF-1 synthesis, sulfate uptake, osteoblast hypertrophy, proliferation of the prechondrocytes, widening of the epiphyseal plate, stimulation of cartilage growth, mineralization and bone remodeling (18, 19). Multiple studies have shown that GH induces growth in two steps: GH directly activates growth hormone receptor (GHR) to stimulate IGF1 synthesis and then IGF-1 acts on target cells to exert some of the physiological effect of GH (2022); thus GH acts directly and indirectly (via IGF-1) on cells of various tissues. This ‘dual effector’ theory of GH action was first postulated by H. Green several decades ago (23). To exert their effects, GH and IGF-1 bind to their cognate receptors [GHR and IGF-1 receptor (IGF-1R)] respectively and activate an array of intracellular signaling systems resulting in the regulation of many genes that mediate cellular differentiation, growth, and other physiological functions (24). In adults, GH induces a rapid fat loss by stimulating lipolysis while inhibiting the lipogenic actions of insulin(25). In addition to JAK-STAT, GH action is also propagated via linkage of GHRs to several cellular effector systems, including Src/SHC, MAPK, PI3K-Akt, and PKC. Differential activation of these pathways under varied physiological conditions determines the anabolic versus catabolic actions of GH (2632). Alterations in GH or GHR action is associated with Laron Syndrome (LS) and Acromegaly in humans.

Recent studies in human adipocytes have shown that GH suppresses the expression of lipid droplet associated protein, fat specific protein 27 (FSP27) and destabilizes Peroxisome Proliferator-Activated Receptor Gamma (PPARγ), the master transcriptional regulator of adipogenesis (3336). This derangement stimulates excessive adipose tissue lipolysis resulting in free fatty acids flux in the systemic circulation. GH-mediated deregulation of fat metabolism, therefore, promotes the progression of insulin resistance, a prime suspect for the development of Type 2 diabetes, cardiovascular and metabolic diseases (3740). Furthermore, GH induces insulin resistance in skeletal muscle and liver via multiple mechanisms to promote its diabetogenic actions (4144). Based on recent in vivo and in vitro studies, this review will provide a concise summary of the molecular mechanisms underlying GH-mediated development of insulin resistance, a pre-requisite for the progression of diabetogenic actions of GH and metabolic diseases.

GH indolence in Dwarf mouse models enhances insulin sensitivity

George Snell first reported dwarfism in mice in 1929 (45). All dwarf mouse models harboring natural spontaneous homozygous mutations (Pit1 mutation in Snell dwarf, Prop-1 mutation in Ames dwarf, a missense mutation in the Growth hormone-releasing hormone in Little mice or targeted gene deletion of Growth hormone receptor gene in Laron dwarf mice) are insulin sensitive, long-lived and metabolically healthy (4652). Interestingly, all these mice share one specific genetic component - impaired GH production/signaling/action. Since GH and insulin are counterregulatory hormones, therefore, reduced insulin secretion, low fasting insulin and glucose levels in conjunction with enhanced insulin sensitivity are the major contributing factors for the improved metabolic health and extended longevity in these dwarf mice (46, 50, 51, 5355). Houssay demonstrated that pituitary insufficiency produces extreme insulin sensitivity in multiple animal models (cat, dog, toad, rabbit, monkey and man) which can be reversed by administration of anterior-pituitary extracts, making GH a prime suspect for its anti-insulinemic effects (56). Since Snell and Ames dwarf mice lack multiple genetic factors, namely growth hormone (GH), prolactin (PRL), and thyroid-stimulating hormone (TSH), the enhanced insulin-sensitive phenotype could not be precisely attributed to a single factor. However, in 1997 targeted disruption of the GH receptor (R) by Kopchick’s laboratory resulted in a mouse model of Laron Syndrome (LS) discussed below (52). These mice are fat and short in stature with low levels of IGF-1 and high levels of GH. Since they lack a functional GHR, they are GH resistant. They also have reduced insulin secretion, display reduced plasma glucose levels, but exhibit increased insulin sensitivity (50, 54). They are also resistant to the development of type 2 diabetes and cancer and are long lived. If fact, they are the longest-lived laboratory mouse (57, 58). This GHR−/− mouse displays a phenotype consistent with patients with LS who possess homozygous inactivation mutations of the GHR gene. These patients are short in stature and ‘chubby’ with low IGF-1, high GH and, again, are GH resistant. Remarkably, they were found to be resistant to diabetes and cancer (59, 60). Therefore, the GHR−/− mouse has helped to precisely pinpoint that GH signaling antagonizes insulin action and disruption of GH action results in low IGF-1 levels as observed in the dwarf mouse models described above. Together, the lack of GH’s direct and indirect effects is a key to their improved metabolic health and very important in their extended longevity. Interestingly, the antagonistic effects of GH and insulin have been conserved during the evolutionary process and are prevalent in many organisms, ranging from yeast to worms and mice to humans (61).

Laron syndrome versus Acromegaly and Insulin sensitivity

Laron syndrome (LS) was first described by Dr. Zvi Laron in 1966. It is a rare genetic disorder inherited in an autosomal recessive manner that is caused by inactivating mutations in the GHR gene and leads to severely depressed IGF-1 levels. LS patients are characterized by dwarfism, facial phenotype, obesity and hypogenitalism (62, 63) with elevated serum GH, but low IGF1 levels, and are GH insensitive or resistant (64, 65). Interestingly, despite having obesity, LS patients are, metabolically healthy, and their risk of developing insulin resistance, cancer, and cardiovascular complications compared to their unaffected relatives is extremely low (59, 60, 6670). Furthermore, Dr. Jamie Guevara-Aguirre’s group reported that an Ecuadorian cohort of LS patients has significantly reduced systemic insulin concentration and a very low HOMA-IR (homeostatic model assessment–insulin resistance) index, indicating higher insulin sensitivity, and the absence of diabetes (59). Hence, anti-insulinemic actions of GH in the GHR−/− mouse mimics that in LS patients.

On the other hand, acromegaly is rare disorder that is characterized by a concomitant increase in both GH and IGF-1. Physiologically, nutrient intake, and glucose suppress GH secretion in healthy subjects but not in patients with acromegaly (71). Chronically elevated levels of GH in these patients induce lipolysis, and hepatic/peripheral insulin resistance (7274) followed by a compensatory increase in insulin secretion to maintain euglycemia (75). Insulin resistance in patients with acromegaly is associated with reduced body fat mass (76, 77). Besides promoting insulin resistance, elevated levels of FFAs can induce corresponding lipotoxicity that can promote beta-cell exhaustion (72, 7881). Therefore, these patients, despite having less fat and more lean mass, if left untreated can develop diabetes, cardiovascular, and other metabolic problems (8285). The first line of treatment of these patients is removal of the GH-secreting pituitary tumors via surgery. If successful, the patients are cured. If remnants of tumor persist, medical treatment is required to decrease GH and/or IGF-1 to normal, age/sex adjusted normal levels at which point insulin sensitivity improves (86).

Interestingly, dwarf mouse models of GH inactivity (Snell, Ames, LS mouse) mimic LS patients and serve an excellent model to study the pathophysiology of GH and its actions in humans. However, recently developed GHR deficient pig model also parallel hallmarks of human LS patients pathophysiology (such as GH insensitivity, high GH and low circulating IGF1 levels, increased adiposity, reduced postnatal body and organ growth) and therefore, offer an appealing model for treatment trials and to nail down the mechanistic studies of human diseases (87). Furthermore, porcine embryonic development resembles that of humans (88), and the pig is closer to human anatomically, physiologically (89) and genetically (90) than the mouse. Finally, the porcine model could bridge the gap between proof-of-concept and discrepancies between rodent and human subject studies (87).

Dawn Phenomenon: A tug of war between GH and Insulin action

Allen Kowarski showed that Type 1 diabetic subjects display a “dawn phenomenon” defined by periodic episodes of hyperglycemia occurring in the early morning hours between 5:00 AM and 8:00 AM (91). This phenomenon reflects an early-morning increase in insulin requirement of patients with insulin-dependent diabetes mellitus (IDDM) as well as Type 2 diabetes patients (92), an effect observed in all age groups but prominently in elderly diabetic subjects (93). In healthy subjects, however, blood glucose and insulin levels remain steady throughout the night, with a slight surge in insulin secretion before dawn to suppress hepatic gluconeogenesis and prevent GH-induced hyperglycemia. The observed increase in insulin in the early morning hours despite stable levels of glucose indicates a temporally increased insulin need in nondiabetic individuals similar to that found in individuals with diabetes (94). Diabetic patients are negatively affected by the dawn phenomenon because of the dysregulated hepatic gluconeogenesis and inability of pancreatic beta-cells to produce compensatory insulin secretion (94, 95). The underlying cause for these hyperglycemic episodes remained a mystery until Gerich’s lab showed that increased plasma glucose levels significantly correlated with peak plasma GH concentration (96).

Interestingly, when nocturnal surges in GH secretion were prevented by infusion of somatostatin, plasma glucose levels and hepatic glucose production were stabilized, demonstrating that nocturnal surges in GH secretion were the prime culprit for dawn phenomenon (96). Further work by Rosenberg’s group showed that GH-induced increase in FFA level contributes to the dawn phenomenon and suppression of sleep-induced GH secretion abolished early morning hyperglycemia (97). Furthermore, GH pulses associated with an increase in endogenous glucose production and decreased rates of peripheral glucose uptake were entirely reversed by pharmacological inhibition of lipolysis with acipimox (98). Therefore, the GH-driven decrease in insulin sensitivity was predominantly due to GH’s effect on adipose tissue lipolysis (99, 100). To our knowledge, the Dawn phenomenon in mice has not been reported, probably due to their nocturnal feeding behavior and unlimited food accessibility.

Growth hormone, hyperinsulinemia and insulin resistance

In healthy human subjects, insulin is released in low-amplitude pulses and rapidly removed from the circulation, possibly to avoid chronic insulin exposure (101, 102). Studies in rats and human subjects in whom pancreatic insulin output was suppressed show that pulsatile delivery of exogenous insulin confers better glycemic control and inhibition of lipolysis over continuous insulin infusion (103, 104). During the development of insulin resistance, the body usually responds by increasing serum insulin concentration to maintain insulin signaling. However, a sustained compensatory increase in insulin concentration causes hyperinsulinemia and dampens insulin response, further making the insulin-resistant state worse (105108). Chronic insulin exposure causes insulin receptor down-regulation and defects in the glucose transport system per se (109). Hyperinsulinemia results in diminished levels of tyrosine and serine phosphorylation of the insulin receptor. Prolonged insulin exposure alters the insulin receptor tyrosine kinase domain and or proteins interacting with this domain in a way that sets the insulin refractory state in the cells (110). Patients with acromegaly display chronically elevated levels of GH and are insulin resistant (111). Due to the antagonistic action of GH on insulin (GH promotes lipolysis while insulin promotes lipogenesis per se), chronically elevated levels of GH in patients with acromegalic are mimicked by a compensatory increase in insulin secretion leading to hyperinsulinemia and therefore, predisposing these patients to insulin resistant/diabetic state (75, 86, 111). Compensatory hyperinsulinemia in acromegalic patients impairs insulin receptor autophosphorylation significantly (110). Insulin binding studies in erythrocytes of the acromegalic patients showed a decreased binding due to a reduction in the receptor number per cell but with no alterations in the affinity of insulin to its receptor (111) and a greater decrease in insulin binding was observed in acromegalic patients with diabetes (112). Similarly, the total insulin receptor concentration per cell was decreased in proportion to the hyperinsulinemia in circulating monocytes, i.e. the receptor concentration was inversely related to the basal level of insulin, similar to that found in patients with obesity, diabetes, and insulin-secreting tumors (113).

Growth hormone and skeletal muscle insulin resistance

GH regulates myofiber number and promotes skeletal muscle development (114). Like adipose tissue, skeletal muscle also stores triglycerides, and to-date, three muscle-specific GHR knockout mouse lines have been generated to study GH function in skeletal muscle. These studies are extensively discussed in a recent review by List et al (58). Muscle-specific deletion of GHR or IGF-1R in mice leads to defective skeletal muscle development and performance (114). Furthermore, mice with skeletal muscle-specific deletion of GHR develop metabolic features, including elevated blood glucose and TG levels, marked peripheral adiposity, insulin resistance, glucose intolerance, and display increased inhibitory phosphorylation of IRS-1 (114). On the contrary, two subsequent studies contradict these findings and show that muscle specific GHRKO mouse lines (mGHRKO and MuGHRKO) show marked health improvements (115). Vijayakumar et al. have shown that skeletal muscle-specific loss of GHR signaling reduced liver and muscle triglyceride content, improved insulin sensitivity, and protected mice against high-fat diet-induced metabolic deterioration (116). List et al. show that male MuGHRKO mice have reduced levels of glucose, insulin, c-peptide, and enhanced glucose tolerance (115), while mGHRKO mice on a high-fat diet have decreased adiposity, inflammation, muscle and hepatic triglyceride content increased carbohydrate utilization, and enhanced insulin sensitivity (116). Furthermore, GH action on muscle had minimal effect on muscle strength or endurance, and disruption of GHR in muscle increased lifespan in male MuGHRKO mice (115). Therefore, it seems that the genetic background of the mouse and the promoter used to drive Cre expression in these mouse lines may be responsible for the contradictory results in these studies. Conversely, transgenic mice overexpressing GH show reduced IR levels and its tyrosine phosphorylation, reduced IRS-1 tyrosine phosphorylation, and defective PI3-kinase activation upon insulin stimulation (42). Similarly, rats treated with GH also show reduced IRS phosphorylation and impaired activation of skeletal muscle insulin signaling due to reduced PI3K activity (42, 117, 118). Furthermore, increased levels of p85α in transgenic mice overexpressing GH or human placental GH, suppress insulin sensitivity in skeletal muscle (42, 119). Interestingly, deletion of a single allele of p85α restored PI3-kinase activity and improved skeletal muscle insulin sensitivity in these mice, suggesting that GH-mediated insulin resistance in skeletal muscle is promoted via its effect on PI3-kinase activity (41).

Although GH replacement therapy (GHRT) in growth hormone deficient (GHD) patients offers clinical benefits, it may deteriorate glucose metabolism in muscles (120), impairs insulin-stimulated glucose turnover into glycolytic flux and glycogen synthesis/glucose storage (121), and promote insulin resistance (122). However, GH-induced acute skeletal muscle insulin resistance can be reversed by pharmacological inhibition of lipolysis or insulin action (123126). GH infusion does not alter Akt (a critical mediator of the insulin signaling pathway that triggers insulin-stimulated glucose uptake via GLUT4(123)) protein expression or GLUT4 protein expression in skeletal muscles of healthy subjects (123), obese (127, 128) or GHRT patients (122). Recent evidence suggests that elevated GH levels as observed under fasting/starvation or upon GH infusion suppress pyruvate dehydrogenase (PDH) activity in muscles (74, 129). This decrease in PDH activity is the result of the metabolic shift from glucose to fatty acid as a primary source of fuel resulting in insulin resistance without altering insulin signaling in human subjects (74, 129). Several studies have shown that FFAs inhibit insulin-stimulated glucose uptake secondary to intramyocellular accumulation of di-acyl glycerol (DAGs) and ceramides, which directly impair insulin signaling (39, 130). Interestingly, GH-induced insulin resistance, despite its dependence on lipolysis, does not affect insulin signaling per se but decreases skeletal muscle glucose uptake (129). Overall, GH exerts skeletal muscle insulin resistance via multiple mechanisms, including increased p85α protein expression (in rodents), reduced IR levels and IRS-1 phosphorylation, decreased PDH activity, and preference for a shift in metabolic fuel from glucose to lipids.

Growth hormone and hepatic insulin resistance

Impaired ability of insulin to suppress hepatic glucose production and adipose lipolysis causes increased hepatic acetyl CoA and links adipose lipolysis to hepatic insulin resistance associated with obesity and T2D (131). Multiple mechanisms contribute to GH-induced hepatic insulin resistance. GH-induced adipose lipolysis alters glucose and lipid oxidation (33, 44), antagonizes insulin action in the liver (44), elicits hepatic gluconeogenesis, lowers peripheral glucose uptake and impairs liver insulin sensitivity in healthy subjects (127, 132, 133). Conversely, low levels of GH associate with hepatic lipid accumulation (134) and GH replacement has been suggested to resolve the fatty liver condition in diet-induced obese rodents and in GH-deficient patients (135). Phosphatase and tensin homolog (PTEN) is a widely known negative regulator of insulin/PI3K signaling (136). Chronic GH treatment upregulates PTEN expression to undermine hepatic insulin signaling (137). GH is thought to increase gluconeogenesis (74) but chronic GH exposure impairs gluconeogenesis (138, 139) and reduces the number of insulin receptors (IR) significantly without altering IR kinase activity in rat liver (140). In agreement with this study, the transgenic mice overexpressing GH also show reduced hepatic IR levels with 3–5 times increase in compensatory phosphorylation of basal IR and IRS-1 and PI3-kinase activity that is refractory/insensitive to further stimulation by insulin (43). Additionally, Akt expression, a downstream target of PI3K, is significantly increased in the liver of GH overexpressing mice to compensate for the defects in the upstream signaling cascade. However, Akt phosphorylation (a measure of insulin sensitivity) was severely diminished, thus resulting in reduced glucose uptake (141). Livers of Ames Dwarf mice and GHR knockout mice on the other hand have impaired GH action/signaling and therefore, exhibit increased IR levels, its cognate phosphorylation and are sensitive to insulin stimulation (142, 143). Furthermore, visceral fat transplant from GHRKO mice into normal mice increases phosphorylated insulin receptor levels and enhances hepatic insulin sensitivity (144). Similarly, blocking GH action/signaling via administration of human GH receptor antagonist, pegvisomant, shows reduction in plasma FFA levels, suppression of hepatic glucose production and improved insulin sensitivity in patients with acromegaly (145) and obese individuals (146). These studies point to the role of cross-organ talk and extrahepatic factors (GH-mediated FFAs and or other unidentified molecular entities of adipogenic origin) in the development of hepatic insulin resistance. Overall, these findings suggest that GH-mediated suppression of insulin’s antilipolytic ability is the prime cause of hepatic insulin resistance (147, 148).

Metabolic effects of GH in Adipose tissue

GH is required for the differentiation of adipocytes and contributes differentially to fat depot development at different anatomical locations (82). Since GH levels are inversely related to adipose tissue mass in both mice and humans, one would expect that GH-mediated reduction of adipose tissue would have health benefits. However, mounting evidence suggests that despite reducing fat mass, GH deteriorates metabolic health in mice and humans (82). Adipose tissue displays significant depot differences with respect to hormonal responsiveness and metabolic activity (149). For example, GHR−/− mice have increased subcutaneous adipose tissue with decreased fibrosis and enhanced insulin sensitivity (33, 150, 151). This may be due to the relative reduction in visceral fat which shows an enhanced lipolysis due to higher β-adrenergic response and reduced α-adrenergic response (152154). GH increases lipolysis in adipocytes partly through the beta-adrenergic response (155). WAT, in response to appropriate stimuli such as cold exposure and β-adrenergic agonist, undergoes browning wherein it acts as an inducible thermogenic adipocyte (beige adipocytes). It increases UCP1 expression and energy production to mimic brown adipose tissue (BAT) function (156159). Therefore, beige adipocytes exhibit UCP1-dependent thermogenesis, and their mitochondria use lipids or carbohydrates as substrates for energy production, similar to brown adipocytes (160). Also, exercise stimulates adrenergic response by potentiating AMP-activated protein kinase (AMPK) to induce PGC1a phosphorylation and activation resulting in mitochondrial biogenesis (161). Chronic exercise, therefore, induces browning of subcutaneous WAT and decreases body weight due to loss of fat mass (162). Interestingly, AMPK subunits and beiging markers such as uncoupling protein 1 (UCP1) and PR domain containing 16 (PRDM16) are upregulated in WAT of transgenic mice overexpressing GH, but downregulated in growth hormone receptor knockout mouse and GH-STAT5 inactivated (GHR-391 mouse) (163). As expected, GHRKO mice and GHR-391 mice are unable to respond to adaptive thermogenesis and other beiging agents (such as ADRB3 agonist treatment and FGF21 infusion), suggesting that in addition to fasting-induced adipose lipolysis, GH also integrates adrenergic input in WAT to promote the formation of beige adipocytes via STAT5 activation (163).

GH contributes to the growth and development during the adolescent years and the GH/IGF-1 axis acts as a prime mediator of insulin resistance across the entire spectrum of normal puberty (164). Furthermore, increased insulin dosage is anticipated with the onset of puberty in patients with insulin-dependent diabetes, and GH exacerbates this process (165168). Low doses of GH-treatment in combination with diet restrictions reduce visceral adipose tissue mass, LDL cholesterol levels, triglycerides, free fatty acids, and improve insulin sensitivity and muscle mass but predisposes individuals to insulin resistance (169, 170). Chronic elevation in GH levels in patients with acromegaly suppresses the anti-lipolytic action of insulin, tipping the balance in favor of fat catabolism resulting in lipotoxicity, which promotes insulin resistance and related metabolic diseases (146, 171).

Patients with elevated levels of GH embody clinical manifestations of the diabetogenic effects of GH since they display increased rate of insulin resistance, hyperinsulinemia, and type 2 diabetes (72, 172, 173). Similarly, patients with acromegaly have elevated level of GH, and therefore, have reduced body fat and increased lean body mass but still develop insulin resistance (74, 174177). Transgenic mice overexpressing GH mimic patients with acromegaly and have less total body fat mass and higher muscle mass than littermate controls but develop insulin resistance (178180). Furthermore, GH administration in GHD children is associated with a redistribution of adipose tissue from an abdominal anatomical location to peripheral location, a significant reduction in abdominal adipocyte size, a significant reduction in overall basal rates of lipogenesis and a reduction in the anti-lipolytic actions of insulin (181). On the contrary, decreased GH action increases visceral as well as total adiposity as observed in LS patients (182) and mouse models of reduced GH action such as Ames dwarf mice (183) and large animal models (87). The decreased adiposity could be attributed to GH-mediated lipolysis and, to a lesser degree, in a preferential increase in intermuscular adipose tissue.

GH exerts anabolic effects through stimulation of IGF-I, insulin, and FFAs under fed conditions. Fasting however, increases GH levels in the systemic circulation, decreases insulin sensitivity, and blunts JAK-STAT pathway activation (184187). Interestingly, the nocturnal increase in FFA during sleep is absent in GH-deficient patients, strongly linking lipolysis to GH (188). Similar to fasting, exercise also induces lipolysis due to adrenergic signaling (189). Elevated levels of GH observed during fasting reduce anti-lipolytic gene G0 switch 2 (G0S2) mRNA and protein expression but increase adipose triglyceride lipase (ATGL, a primary lipase in adipose tissue), protein activity with a concomitant rise in circulating fatty acids (190). However, both ATGL and G0S2 remain unchanged during exercise, suggesting that the ATGL-G0S2 complex is an important long-term regulator of lipolysis under physiological conditions (such as fasting) in humans (191). Recently, we have shown that GH downregulates expression of the negative regulators of lipolysis, namely, FSP27 and G0S2 (34, 36). Forced expression of FSP27 however, suppresses GH-induced adipose lipolysis and restores insulin sensitivity (34, 36). As stated above, studies in human subjects and mouse models show that GH affects adipose insulin sensitivity via multiple mechanisms including increased stimulation of adipose lipolysis (192, 193), expression of p85α in mice (194), increased activity of hormone-sensitive lipase (HSL) and ATGL in adipose tissue (36, 195198), reduced insulin receptor expression (139), reduced adipose tissue lipoprotein lipase (LPL) activity, and impaired expression of FSP27 and G0S2 (34, 36) discussed below and shown in Figure 1. Overall, these studies provide a solid foundation and framework and support that GH-mediated lipolysis is one of the critical regulators of insulin action in both healthy subjects and diabetic patients.

Figure 1. Intracellular model of GH signaling under normal and pathological conditions in Human adipocytes:

Figure 1.

Upon stimulation by GH binding, GHR activates Jak2. Activated Jak2 phosphorylates GHR on tyrosine residues which in turn recruit members of the STAT family of transcription factors. Additionally, Src kinase and Shc adaptor proteins are also recruited to the GHR to activate a series of signaling pathways. STATs homodimerize or heterodimerize and translocate to the nucleus to regulate an array of genes under physiological conditions. Coordinated action of STATs and other signaling pathways leads to a balanced state of physiological lipolysis to provide energy for growth, development and metabolism under normal and fasting/starvation conditions.

However, chronic levels of elevated GH (as seen in patients with acromegaly) leads to activation of the Ras-Raf-MEK-ERK pathway which dominates other signaling pathways. As a result, phopshorylated ERK translocates to nucleus and phosphorylates PPARγ. Phopshorylated PPARγ is targeted for degradation leading to downregulation of anti-lipolytic genes such as FSP27 and G0S2. Sustained levels of systemic FFAs as a result of deregulated lipolysis cause lipotoxicity and pave the way for the development of insulin resistance, diabetes and related metabolic diseases.

Transcriptional Regulation of Lipid droplet (LD) associated Proteins and mechanisms of GH-induced lipolysis

Fatty acids are stored as triglycerides in LDs in adipocytes. LDs contain various proteins on their surface (199), which are involved in lipogenesis, lipolysis, or maintaining lipid homeostasis. LD-associated proteins regulating lipolysis such as ATGL, G0S2, PLIN1, and FSP27, also known as Cell Death-Inducing DFFA-like Effector Protein C (CIDEC) are all direct transcriptional targets of master adipogenesis regulator PPARγ (200202). PPARγ can sense and interpret fatty acid signals (203, 204). Therefore, alterations in the intracellular concentration of FFAs directly impact PPARγ-dependent gene regulation (205). Recent studies have highlighted the role of FSP27 as a GH target to induce lipolysis and insulin resistance (3336). FSP27 is highly expressed in WAT and regulates lipid droplet dynamics and fat metabolism (206212). Puri’s lab has recently described its major role in adipocyte lipolysis and insulin sensitivity in human adipocytes (206, 207).

Recent studies in human subjects, primary human adipocytes and 3T3-L1 adipocytes have shown that GH treatment downregulates FSP27 and G0S2 expression, resulting in LD fragmentation and systemic flux of FFAs (34, 36). Insulin decrease lipolysis via regulating FSP27 expression. FSP27 interacts with ATGL to regulate its lipolytic action in human adipocytes (206). Also, upon insulin stimulation, FSP27 recruits EGR1 at the ATGL promoter to directly inhibit ATGL transcription and its lipolytic capacity, suggesting that FSP27 acts at multiple levels to tightly regulate ATGL-mediated lipolysis (213). Mechanistically, GH binding to its receptor activates multiple signaling pathways, including the Ras-Raf-MEK-ERK1,2 pathway (26, 214216). MEK1-mediated phosphorylation of PPARγ leads to its nuclear export and degradation (217, 218). GH-mediated activation of MEK/ERK signaling downregulates FSP27, destabilizes PPARγ, and increases HSL phosphorylation to promote lipolysis in human adipocytes (36). Forced expression of FSP27 or blocking MEK/ERK signaling stabilized PPARγ in the nucleus, improved insulin signaling and restored GH-induced lipolysis to normal levels (34, 36). Counterintuitively, GH administration also resulted in FSP27 mRNA upregulation suggestive of an additional transcription regulation mechanism for FSP27 expression (34). Such a mechanism is physiologically obligatory to optimally regulate lipolysis and maintain adipose tissue homeostasis. GH also signals through STAT5, which can homodimerize or heterodimerize with other STAT family members and translocate to the nucleus in MEK and PPARγ independent manner to regulate gene expression and mediate GH effects (26). Interestingly, inhibiting STAT5 phosphorylation with a subsequent GH stimulation nearly abolishes FSP27 mRNA expression in 3T3-L1 adipocytes (34). These results suggest that GH-mediated STAT5 phosphorylation upregulates FSP27 expression under physiological conditions to keep GH stimulated MEK/ERK signaling under check and maintain an optimal level of lipolysis. Furthermore, FSP27 is significantly reduced in the perigonadal fat and subcutaneous fat of STAT5ΔN/ΔN-mutant mice, which express hypomorphic forms of both Stat5a and Stat5b (34). These studies provide molecular insights as to how GH-mediated activation of MEK/ERK causes destabilization of the transcription factor PPARγ resulting in the downregulation of FSP27, G0S2, and other lipid-droplet associated genes while simultaneously phosphorylating STAT5 to regulate LD homeostasis and prevent excessive lipolysis.

Concluding remarks

GH and insulin both have mitogenic and metabolic effects (Figure 2). However, whether GH and insulin actions will synergize to promote mitogenic or antagonize to promote metabolic effects, depends on the duration of exposure to the respective hormone. Multiple lines of evidence suggest that impaired adipose tissue homeostasis plays a significant role in maintaining whole-body insulin sensitivity, and GH-mediated adipose tissue lipolysis is one of the most critical regulators of insulin resistance in both healthy subjects and diabetic patients.

Figure 2. Synergistic and antagonistic effects of growth hormone and insulin signaling.

Figure 2.

Acute GH and insulin exposure act synergistically (as shown in green) to promote normal growth, metabolism, differentiation, and development. However, chronic GH exposure antagonizes insulin action and vice-versa (shown by red arrows) to promote insulin resistance in multiple organs (Liver, skeletal muscle, and adipose tissue) via multiple mechanisms. Dysregulated GH and insulin action, under pathological conditions alter gene/protein expression and their enzymatic activity in insulin sensitive organs to promote insulin resistance that leads to the development of Type 2 diabetes and related metabolic diseases.

Acknowledgments:

I would like to thank my colleagues, Drs. Mark Slayton, Bijinu Balakrishnan, Abhishek Gupta, Edward List, and Yanrong Qian for their help, and critical comments. I sincerely apologize to those whose work, although pertinent to this topic, was not mentioned due to inadvertent omission. This work was supported by NIH grants RO1DK101711, R01HL140836, RO1DK124126, and R01MD012579 to VP, and funds from Osteopathic Heritage Foundation’s Vision 2020 to Heritage college of Osteopathic medicine at Ohio University.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References:

  • 1.Evans HM, Long JA. Characteristic Effects upon Growth, Oestrus and Ovulation Induced by the Intraperitoneal Administration of Fresh Anterior Hypophyseal Substance. Proc Natl Acad Sci U S A. 1922;8(3):38–9. doi: 10.1073/pnas.8.3.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Biasotti A, Houssay BA. Phlorrhizin diabetes in fasting or fed hypophysectomized dogs. J Physiol. 1932;77(1):81–91. doi: 10.1113/jphysiol.1932.sp002952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Young FG. The diabetogenic action of crude anterior pituitary extracts. Biochem J. 1938;32(3):513–23. doi: 10.1042/bj0320513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Young FG. “Growth” and the Diabetogenic Action of Anterior Pituitary Preparations. Br Med J. 1941;2(4225):897–901. doi: 10.1136/bmj.2.4225.897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Young FG. Growth and diabetes in normal animals treated with pituitary (anterior lobe) diabetogenic extract. Biochem J. 1945;39(5):515–36. doi: 10.1042/bj0390515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Houssay BA, Foglia VG, Smyth FS, Rietti CT, Houssay AB. The Hypophysis and Secretion of Insulin. J Exp Med. 1942;75(5):547–66. doi: 10.1084/jem.75.5.547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Li CH, Evans HM. The Isolation of Pituitary Growth Hormone. Science. 1944;99(2566):183–4. doi: 10.1126/science.99.2566.183. [DOI] [PubMed] [Google Scholar]
  • 8.Bornstein J, Reid E, Young FG. The hyperglycaemic action of blood from animals treated with growth hormone. Nature. 1951;168(4282):903–5. doi: 10.1038/168903a0. [DOI] [PubMed] [Google Scholar]
  • 9.Cotes PM, Reid E, Young FG. Diabetogenic action of pure anterior pituitary growth hormone. Nature. 1949;164(4162):209–11. doi: 10.1038/164209a0. [DOI] [PubMed] [Google Scholar]
  • 10.Houssay BA, Rodriguez RR. Diabetogenic action of different preparations of growth hormone. Endocrinology. 1953;53(1):114–6. doi: 10.1210/endo-53-1-114. [DOI] [PubMed] [Google Scholar]
  • 11.Houssay BA, Rodriguez RR, Cardeza AF. [Diabetogenic action of the pituitary growth hormone]. C R Seances Soc Biol Fil. 1954;148(9–10):910–1. [PubMed] [Google Scholar]
  • 12.Rabinowitz D, Klassen GA, Zierler KL. Effect of Human Growth Hormone on Muscle and Adipose Tissue Metabolism in the Forearm of Man. J Clin Invest. 1965;44:51–61. doi: 10.1172/JCI105126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ranke MB, Wit JM. Growth hormone - past, present and future. Nat Rev Endocrinol. 2018;14(5):285–300. doi: 10.1038/nrendo.2018.22. [DOI] [PubMed] [Google Scholar]
  • 14.Le Roith D, Bondy C, Yakar S, Liu JL, Butler A. The somatomedin hypothesis: 2001. Endocr Rev. 2001;22(1):53–74. doi: 10.1210/edrv.22.1.0419. [DOI] [PubMed] [Google Scholar]
  • 15.Ohlsson C, Nilsson A, Isaksson O, Lindahl A. Growth hormone induces multiplication of the slowly cycling germinal cells of the rat tibial growth plate. Proc Natl Acad Sci U S A. 1992;89(20):9826–30. doi: 10.1073/pnas.89.20.9826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kastin AJ, Redding TW, Hall R, Besser GM, Schally AV. Lipid mobilizing hormones of the hypothalamus and pituitary. Pharmacol Biochem Behav. 1975;3(1 Suppl):121–6. [PubMed] [Google Scholar]
  • 17.Singhal V, Goh BC, Bouxsein ML, Faugere MC, DiGirolamo DJ. Osteoblast-restricted Disruption of the Growth Hormone Receptor in Mice Results in Sexually Dimorphic Skeletal Phenotypes. Bone Res. 2013;1(1):85–97. doi: 10.4248/BR201301006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kaplan SA, Cohen P. The somatomedin hypothesis 2007: 50 years later. J Clin Endocrinol Metab. 2007;92(12):4529–35. doi: 10.1210/jc.2007-0526. [DOI] [PubMed] [Google Scholar]
  • 19.Ohlsson C, Bengtsson BA, Isaksson OG, Andreassen TT, Slootweg MC. Growth hormone and bone. Endocr Rev. 1998;19(1):55–79. doi: 10.1210/edrv.19.1.0324. [DOI] [PubMed] [Google Scholar]
  • 20.Nilsson A, Carlsson B, Isgaard J, Isaksson OG, Rymo L. Regulation by GH of insulin-like growth factor-I mRNA expression in rat epiphyseal growth plate as studied with in-situ hybridization. J Endocrinol. 1990;125(1):67–74. doi: 10.1677/joe.0.1250067. [DOI] [PubMed] [Google Scholar]
  • 21.Nilsson A, Isgaard J, Lindahl A, Dahlstrom A, Skottner A, Isaksson OG. Regulation by growth hormone of number of chondrocytes containing IGF-I in rat growth plate. Science. 1986;233(4763):571–4. doi: 10.1126/science.3523759. [DOI] [PubMed] [Google Scholar]
  • 22.Nilsson A, Lindahl A, Eden S, Isaksson OG. Demonstration of growth hormone receptors in cultured rat epiphyseal chondrocytes by specific binding of growth hormone and immunohistochemistry. J Endocrinol. 1989;122(1):69–77. doi: 10.1677/joe.0.1220069. [DOI] [PubMed] [Google Scholar]
  • 23.Green H, Morikawa M, Nixon T. A dual effector theory of growth-hormone action. Differentiation. 1985;29(3):195–8. doi: 10.1111/j.1432-0436.1985.tb00316.x. [DOI] [PubMed] [Google Scholar]
  • 24.Yakar S, Isaksson O. Regulation of skeletal growth and mineral acquisition by the GH/IGF-1 axis: Lessons from mouse models. Growth Horm IGF Res. 2016;28:26–42. doi: 10.1016/j.ghir.2015.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Veldhuis JD, Roemmich JN, Richmond EJ, Rogol AD, Lovejoy JC, Sheffield-Moore M, Mauras N, Bowers CY. Endocrine control of body composition in infancy, childhood, and puberty. Endocr Rev. 2005;26(1):114–46. doi: 10.1210/er.2003-0038. [DOI] [PubMed] [Google Scholar]
  • 26.Brooks AJ, Waters MJ. The growth hormone receptor: mechanism of activation and clinical implications. Nat Rev Endocrinol. 2010;6(9):515–25. doi: 10.1038/nrendo.2010.123. [DOI] [PubMed] [Google Scholar]
  • 27.Dinerstein-Cali H, Ferrag F, Kayser C, Kelly PA, Postel-Vinay M. Growth hormone (GH) induces the formation of protein complexes involving Stat5, Erk2, Shc and serine phosphorylated proteins. Mol Cell Endocrinol. 2000;166(2):89–99. doi: 10.1016/s0303-7207(00)00277-x. [DOI] [PubMed] [Google Scholar]
  • 28.Lanning NJ, Carter-Su C. Recent advances in growth hormone signaling. Rev Endocr Metab Disord. 2006;7(4):225–35. doi: 10.1007/s11154-007-9025-5. [DOI] [PubMed] [Google Scholar]
  • 29.Love DW, Whatmore AJ, Clayton PE, Silva CM. Growth hormone stimulation of the mitogen-activated protein kinase pathway is cell type specific. Endocrinology. 1998;139(4):1965–71. doi: 10.1210/endo.139.4.5884. [DOI] [PubMed] [Google Scholar]
  • 30.Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 2001;414(6865):799–806. doi: 10.1038/414799a. [DOI] [PubMed] [Google Scholar]
  • 31.Thirone AC, Carvalho CR, Saad MJ. Growth hormone stimulates the tyrosine kinase activity of JAK2 and induces tyrosine phosphorylation of insulin receptor substrates and Shc in rat tissues. Endocrinology. 1999;140(1):55–62. doi: 10.1210/endo.140.1.6417. [DOI] [PubMed] [Google Scholar]
  • 32.Zhu T, Ling L, Lobie PE. Identification of a JAK2-independent pathway regulating growth hormone (GH)-stimulated p44/42 mitogen-activated protein kinase activity. GH activation of Ral and phospholipase D is Src-dependent. J Biol Chem. 2002;277(47):45592–603. doi: 10.1074/jbc.M201385200. [DOI] [PubMed] [Google Scholar]
  • 33.Kopchick JJ, Berryman DE, Puri V, Lee KY, Jorgensen JOL. The effects of growth hormone on adipose tissue: old observations, new mechanisms. Nat Rev Endocrinol. 2020;16(3):135–46. doi: 10.1038/s41574-019-0280-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sharma R, Luong Q, Sharma VM, Harberson M, Harper B, Colborn A, Berryman DE, Jessen N, Jorgensen JOL, Kopchick JJ, Puri V, Lee KY. Growth hormone controls lipolysis by regulation of FSP27 expression. J Endocrinol. 2018;239(3):289–301. doi: 10.1530/JOE-18-0282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sharma VM. Emerging Mechanisms of GH-Induced Lipolysis and Insulin Resistance. Pediatr Endocrinol Rev. 2019;17(1):4–16. doi: 10.17458/per.vol17.2019.s.ghlipolysisandinsulinresistance. [DOI] [PubMed] [Google Scholar]
  • 36.Sharma VM, Vestergaard ET, Jessen N, Kolind-Thomsen P, Nellemann B, Nielsen TS, Vendelbo MH, Moller N, Sharma R, Lee KY, Kopchick JJ, Jorgensen JOL, Puri V. Growth hormone acts along the PPARgamma-FSP27 axis to stimulate lipolysis in human adipocytes. Am J Physiol Endocrinol Metab. 2019;316(1):E34–E42. doi: 10.1152/ajpendo.00129.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Reaven G Insulin resistance and coronary heart disease in nondiabetic individuals. Arterioscler Thromb Vasc Biol. 2012;32(8):1754–9. doi: 10.1161/ATVBAHA.111.241885. [DOI] [PubMed] [Google Scholar]
  • 38.Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and insulin resistance: underlying causes and modification by exercise training. Compr Physiol. 2013;3(1):1–58. doi: 10.1002/cphy.c110062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell. 2012;148(5):852–71. doi: 10.1016/j.cell.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yazici D, Sezer H. Insulin Resistance, Obesity and Lipotoxicity. Adv Exp Med Biol. 2017;960:277–304. doi: 10.1007/978-3-319-48382-5_12. [DOI] [PubMed] [Google Scholar]
  • 41.Barbour LA, Mizanoor Rahman S, Gurevich I, Leitner JW, Fischer SJ, Roper MD, Knotts TA, Vo Y, McCurdy CE, Yakar S, Leroith D, Kahn CR, Cantley LC, Friedman JE, Draznin B. Increased P85alpha is a potent negative regulator of skeletal muscle insulin signaling and induces in vivo insulin resistance associated with growth hormone excess. J Biol Chem. 2005;280(45):37489–94. doi: 10.1074/jbc.M506967200. [DOI] [PubMed] [Google Scholar]
  • 42.Dominici FP, Cifone D, Bartke A, Turyn D. Alterations in the early steps of the insulin-signaling system in skeletal muscle of GH-transgenic mice. Am J Physiol. 1999;277(3):E447–54. doi: 10.1152/ajpendo.1999.277.3.E447. [DOI] [PubMed] [Google Scholar]
  • 43.Dominici FP, Cifone D, Bartke A, Turyn D. Loss of sensitivity to insulin at early events of the insulin signaling pathway in the liver of growth hormone-transgenic mice. J Endocrinol. 1999;161(3):383–92. doi: 10.1677/joe.0.1610383. [DOI] [PubMed] [Google Scholar]
  • 44.Piatti PM, Monti LD, Caumo A, Conti M, Magni F, Galli-Kienle M, Fochesato E, Pizzini A, Baldi L, Valsecchi G, Pontiroli AE. Mediation of the hepatic effects of growth hormone by its lipolytic activity. J Clin Endocrinol Metab. 1999;84(5):1658–63. doi: 10.1210/jcem.84.5.5685. [DOI] [PubMed] [Google Scholar]
  • 45.Snell GD. Dwarf, a New Mendelian Recessive Character of the House Mouse. Proc Natl Acad Sci U S A. 1929;15(9):733–4. doi: 10.1073/pnas.15.9.733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, 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. 2003;144(9):3799–810. doi: 10.1210/en.2003-0374. [DOI] [PubMed] [Google Scholar]
  • 47.Eicher EM, Beamer WG. Inherited ateliotic dwarfism in mice. Characteristics of the mutation, little, on chromosome 6. J Hered. 1976;67(2):87–91. doi: 10.1093/oxfordjournals.jhered.a108682. [DOI] [PubMed] [Google Scholar]
  • 48.Flurkey K, Papaconstantinou J, Miller RA, 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. 2001;98(12):6736–41. doi: 10.1073/pnas.111158898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Godfrey P, Rahal JO, Beamer WG, Copeland NG, Jenkins NA, Mayo KE. GHRH receptor of little mice contains a missense mutation in the extracellular domain that disrupts receptor function. Nat Genet. 1993;4(3):227–32. doi: 10.1038/ng0793-227. [DOI] [PubMed] [Google Scholar]
  • 50.List EO, Sackmann-Sala L, Berryman DE, Funk K, Kelder B, Gosney ES, Okada S, Ding J, Cruz-Topete D, Kopchick JJ. Endocrine parameters and phenotypes of the growth hormone receptor gene disrupted (GHR−/−) mouse. Endocr Rev. 2011;32(3):356–86. doi: 10.1210/er.2010-0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sun LY, Spong A, Swindell WR, Fang Y, Hill C, Huber JA, Boehm JD, Westbrook R, Salvatori R, Bartke A. Growth hormone-releasing hormone disruption extends lifespan and regulates response to caloric restriction in mice. Elife. 2013;2:e01098. doi: 10.7554/eLife.01098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhou Y, Xu BC, Maheshwari HG, He L, Reed M, Lozykowski M, Okada S, Cataldo L, Coschigamo K, Wagner TE, Baumann G, Kopchick JJ. A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci U S A. 1997;94(24):13215–20. doi: 10.1073/pnas.94.24.13215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bartke A Single-gene mutations and healthy ageing in mammals. Philos Trans R Soc Lond B Biol Sci. 2011;366(1561):28–34. doi: 10.1098/rstb.2010.0281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bartke A, Brown-Borg H. Life extension in the dwarf mouse. Curr Top Dev Biol. 2004;63:189–225. doi: 10.1016/S0070-2153(04)63006-7. [DOI] [PubMed] [Google Scholar]
  • 55.Wiesenborn DS, Ayala JE, King E, Masternak MM. Insulin sensitivity in long-living Ames dwarf mice. Age (Dordr). 2014;36(5):9709. doi: 10.1007/s11357-014-9709-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.H BA. THE HYPOPHYSIS AND METABOLISM. The New England Journal of Medicine. 1936;214(20):961–71. doi: 10.1056/NEJM193605142142001. [DOI] [Google Scholar]
  • 57.H P. Money for old mice. Nature. 2003. doi: 10.1038/news030915-13. [DOI] [Google Scholar]
  • 58.List EO, Berryman DE, Jensen EA, Kulkarni P, McKenna S, Kopchick JJ. New insights of growth hormone (GH) actions from tissue-specific GH receptor knockouts in mice. Arch Endocrinol Metab. 2019;63(6):557–67. doi: 10.20945/2359-3997000000185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, Martin-Montalvo A, Saavedra J, Ingles S, de Cabo R, Cohen P, Longo VD. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3(70):70ra13. doi: 10.1126/scitranslmed.3001845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Steuerman R, Shevah O, Laron Z. Congenital IGF1 deficiency tends to confer protection against post-natal development of malignancies. Eur J Endocrinol. 2011;164(4):485–9. doi: 10.1530/EJE-10-0859. [DOI] [PubMed] [Google Scholar]
  • 61.Barbieri M, Bonafe M, Franceschi C, Paolisso G. Insulin/IGF-I-signaling pathway: an evolutionarily conserved mechanism of longevity from yeast to humans. Am J Physiol Endocrinol Metab. 2003;285(5):E1064–71. doi: 10.1152/ajpendo.00296.2003. [DOI] [PubMed] [Google Scholar]
  • 62.Janecka A, Kolodziej-Rzepa M, Biesaga B. Clinical and Molecular Features of Laron Syndrome, A Genetic Disorder Protecting from Cancer. In Vivo. 2016;30(4):375–81. [PubMed] [Google Scholar]
  • 63.Laron Z, Pertzelan A, Mannheimer S. Genetic pituitary dwarfism with high serum concentation of growth hormone--a new inborn error of metabolism? Isr J Med Sci. 1966;2(2):152–5. [PubMed] [Google Scholar]
  • 64.Eshet R, Laron Z, Pertzelan A, Arnon R, Dintzman M. Defect of human growth hormone receptors in the liver of two patients with Laron-type dwarfism. Isr J Med Sci. 1984;20(1):8–11. [PubMed] [Google Scholar]
  • 65.Godowski PJ, Leung DW, Meacham LR, Galgani JP, Hellmiss R, Keret R, Rotwein PS, Parks JS, Laron Z, Wood WI. Characterization of the human growth hormone receptor gene and demonstration of a partial gene deletion in two patients with Laron-type dwarfism. Proc Natl Acad Sci U S A. 1989;86(20):8083–7. doi: 10.1073/pnas.86.20.8083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Guevara-Aguirre J, Rosenbloom AL, Balasubramanian P, Teran E, Guevara-Aguirre M, Guevara C, Procel P, Alfaras I, De Cabo R, Di Biase S, Narvaez L, Saavedra J, Longo VD. GH Receptor Deficiency in Ecuadorian Adults Is Associated With Obesity and Enhanced Insulin Sensitivity. J Clin Endocrinol Metab. 2015;100(7):2589–96. doi: 10.1210/jc.2015-1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Laron Z Do deficiencies in growth hormone and insulin-like growth factor-1 (IGF-1) shorten or prolong longevity? Mech Ageing Dev. 2005;126(2):305–7. doi: 10.1016/j.mad.2004.08.022. [DOI] [PubMed] [Google Scholar]
  • 68.Laron Z, Kauli R. Fifty seven years of follow-up of the Israeli cohort of Laron Syndrome patients-From discovery to treatment. Growth Horm IGF Res. 2016;28:53–6. doi: 10.1016/j.ghir.2015.08.004. [DOI] [PubMed] [Google Scholar]
  • 69.Samaras TT, Elrick H. Height, body size and longevity. Acta Med Okayama. 1999;53(4):149–69. [PubMed] [Google Scholar]
  • 70.Shevah O, Laron Z. Patients with congenital deficiency of IGF-I seem protected from the development of malignancies: a preliminary report. Growth Horm IGF Res. 2007;17(1):54–7. doi: 10.1016/j.ghir.2006.10.007. [DOI] [PubMed] [Google Scholar]
  • 71.Earll JM, Sparks LL, Forsham PH. Glucose suppression of serum growth hormone in the diagnosis of acromegaly. JAMA. 1967;201(8):628–30. [PubMed] [Google Scholar]
  • 72.Hansen I, Tsalikian E, Beaufrere B, Gerich J, Haymond M, Rizza R. Insulin resistance in acromegaly: defects in both hepatic and extrahepatic insulin action. Am J Physiol. 1986;250(3 Pt 1):E269–73. doi: 10.1152/ajpendo.1986.250.3.E269. [DOI] [PubMed] [Google Scholar]
  • 73.Karlander S, Vranic M, Efendic S. Increased glucose turnover and glucose cycling in acromegalic patients with normal glucose tolerance. Diabetologia. 1986;29(11):778–83. doi: 10.1007/BF00873216. [DOI] [PubMed] [Google Scholar]
  • 74.Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev. 2009;30(2):152–77. doi: 10.1210/er.2008-0027. [DOI] [PubMed] [Google Scholar]
  • 75.Kasayama S, Otsuki M, Takagi M, Saito H, Sumitani S, Kouhara H, Koga M, Saitoh Y, Ohnishi T, Arita N. Impaired beta-cell function in the presence of reduced insulin sensitivity determines glucose tolerance status in acromegalic patients. Clin Endocrinol (Oxf). 2000;52(5):549–55. doi: 10.1046/j.1365-2265.2000.00986.x. [DOI] [PubMed] [Google Scholar]
  • 76.Reyes-Vidal CM, Mojahed H, Shen W, Jin Z, Arias-Mendoza F, Fernandez JC, Gallagher D, Bruce JN, Post KD, Freda PU. Adipose Tissue Redistribution and Ectopic Lipid Deposition in Active Acromegaly and Effects of Surgical Treatment. J Clin Endocrinol Metab. 2015;100(8):2946–55. doi: 10.1210/jc.2015-1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Winhofer Y, Wolf P, Krssak M, Wolfsberger S, Tura A, Pacini G, Gessl A, Raber W, Kukurova IJ, Kautzky-Willer A, Knosp E, Trattnig S, Krebs M, Luger A. No evidence of ectopic lipid accumulation in the pathophysiology of the acromegalic cardiomyopathy. J Clin Endocrinol Metab. 2014;99(11):4299–306. doi: 10.1210/jc.2014-2242. [DOI] [PubMed] [Google Scholar]
  • 78.Bollheimer LC, Skelly RH, Chester MW, McGarry JD, Rhodes CJ. Chronic exposure to free fatty acid reduces pancreatic beta cell insulin content by increasing basal insulin secretion that is not compensated for by a corresponding increase in proinsulin biosynthesis translation. J Clin Invest. 1998;101(5):1094–101. doi: 10.1172/JCI420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Maedler K, Oberholzer J, Bucher P, Spinas GA, Donath MY. Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose on human pancreatic beta-cell turnover and function. Diabetes. 2003;52(3):726–33. doi: 10.2337/diabetes.52.3.726. [DOI] [PubMed] [Google Scholar]
  • 80.Maedler K, Spinas GA, Dyntar D, Moritz W, Kaiser N, Donath MY. Distinct effects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes. 2001;50(1):69–76. doi: 10.2337/diabetes.50.1.69. [DOI] [PubMed] [Google Scholar]
  • 81.McGarry JD, Dobbins RL. Fatty acids, lipotoxicity and insulin secretion. Diabetologia. 1999;42(2):128–38. doi: 10.1007/s001250051130. [DOI] [PubMed] [Google Scholar]
  • 82.Berryman DE, List EO. Growth Hormone’s Effect on Adipose Tissue: Quality versus Quantity. Int J Mol Sci. 2017;18(8). doi: 10.3390/ijms18081621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Frara S, Maffezzoni F, Mazziotti G, Giustina A. Current and Emerging Aspects of Diabetes Mellitus in Acromegaly. Trends Endocrinol Metab. 2016;27(7):470–83. doi: 10.1016/j.tem.2016.04.014. [DOI] [PubMed] [Google Scholar]
  • 84.Hannon AM, Thompson CJ, Sherlock M. Diabetes in Patients With Acromegaly. Curr Diab Rep. 2017;17(2):8. doi: 10.1007/s11892-017-0838-7. [DOI] [PubMed] [Google Scholar]
  • 85.Liu C, Wang J, Li J, Zeng M, Luo R, Shen J, Sun X, Han W, Wang L. Synthesis of N-Doped Hollow-Structured Mesoporous Carbon Nanospheres for High-Performance Supercapacitors. ACS Appl Mater Interfaces. 2016;8(11):7194–204. doi: 10.1021/acsami.6b02404. [DOI] [PubMed] [Google Scholar]
  • 86.Roelfsema F, Frolich M. Glucose tolerance and plasma immunoreactive insulin levels in acromegalics before and after selective transsphenoidal surgery. Clin Endocrinol (Oxf). 1985;22(4):531–7. doi: 10.1111/j.1365-2265.1985.tb00153.x. [DOI] [PubMed] [Google Scholar]
  • 87.Hinrichs A, Kessler B, Kurome M, Blutke A, Kemter E, Bernau M, Scholz AM, Rathkolb B, Renner S, Bultmann S, Leonhardt H, de Angelis MH, Nagashima H, Hoeflich A, Blum WF, Bidlingmaier M, Wanke R, Dahlhoff M, Wolf E. Growth hormone receptor-deficient pigs resemble the pathophysiology of human Laron syndrome and reveal altered activation of signaling cascades in the liver. Mol Metab. 2018;11:113–28. doi: 10.1016/j.molmet.2018.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kobayashi T, Zhang H, Tang WWC, Irie N, Withey S, Klisch D, Sybirna A, Dietmann S, Contreras DA, Webb R, Allegrucci C, Alberio R, Surani MA. Principles of early human development and germ cell program from conserved model systems. Nature. 2017;546(7658):416–20. doi: 10.1038/nature22812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Walters EM, Prather RS. Advancing swine models for human health and diseases. Mo Med. 2013;110(3):212–5. [PMC free article] [PubMed] [Google Scholar]
  • 90.Wernersson R, Schierup MH, Jorgensen FG, Gorodkin J, Panitz F, Staerfeldt HH, Christensen OF, Mailund T, Hornshoj H, Klein A, Wang J, Liu B, Hu S, Dong W, Li W, Wong GK, Yu J, Wang J, Bendixen C, Fredholm M, Brunak S, Yang H, Bolund L. Pigs in sequence space: a 0.66X coverage pig genome survey based on shotgun sequencing. BMC Genomics. 2005;6:70. doi: 10.1186/1471-2164-6-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Schmidt MI, Hadji-Georgopoulos A, Rendell M, Margolis S, Kowarski A. The dawn phenomenon, an early morning glucose rise: implications for diabetic intraday blood glucose variation. Diabetes Care. 1981;4(6):579–85. doi: 10.2337/diacare.4.6.579. [DOI] [PubMed] [Google Scholar]
  • 92.Bolli GB, Gerich JE. The “dawn phenomenon”--a common occurrence in both non-insulin-dependent and insulin-dependent diabetes mellitus. N Engl J Med. 1984;310(12):746–50. doi: 10.1056/NEJM198403223101203. [DOI] [PubMed] [Google Scholar]
  • 93.Monnier L, Colette C, Sardinoux M, Baptista G, Regnier-Zerbib A, Owens D. Frequency and severity of the dawn phenomenon in type 2 diabetes: relationship to age. Diabetes Care. 2012;35(12):2597–9. doi: 10.2337/dc12-0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Schmidt MI, Lin QX, Gwynne JT, Jacobs S. Fasting early morning rise in peripheral insulin: evidence of the dawn phenomenon in nondiabetes. Diabetes Care. 1984;7(1):32–5. doi: 10.2337/diacare.7.1.32. [DOI] [PubMed] [Google Scholar]
  • 95.Rybicka M, Krysiak R, Okopien B. The dawn phenomenon and the Somogyi effect - two phenomena of morning hyperglycaemia. Endokrynol Pol. 2011;62(3):276–84. [PubMed] [Google Scholar]
  • 96.Campbell PJ, Bolli GB, Cryer PE, Gerich JE. Pathogenesis of the dawn phenomenon in patients with insulin-dependent diabetes mellitus. Accelerated glucose production and impaired glucose utilization due to nocturnal surges in growth hormone secretion. N Engl J Med. 1985;312(23):1473–9. doi: 10.1056/NEJM198506063122302. [DOI] [PubMed] [Google Scholar]
  • 97.Davidson MB, Harris MD, Ziel FH, Rosenberg CS. Suppression of sleep-induced growth hormone secretion by anticholinergic agent abolishes dawn phenomenon. Diabetes. 1988;37(2):166–71. doi: 10.2337/diab.37.2.166. [DOI] [PubMed] [Google Scholar]
  • 98.Nielsen S, Moller N, Pedersen SB, Christiansen JS, Jorgensen JO. The effect of long-term pharmacological antilipolysis on substrate metabolism in growth hormone (GH)-substituted GH-deficient adults. J Clin Endocrinol Metab. 2002;87(7):3274–8. doi: 10.1210/jcem.87.7.8597. [DOI] [PubMed] [Google Scholar]
  • 99.Salgin B, Marcovecchio ML, Williams RM, Jackson SJ, Bluck LJ, Humphreys SM, Acerini CL, Dunger DB. Effects of growth hormone and free fatty acids on insulin sensitivity in patients with type 1 diabetes. J Clin Endocrinol Metab. 2009;94(9):3297–305. doi: 10.1210/jc.2009-0378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Segerlantz M, Bramnert M, Manhem P, Laurila E, Groop LC. Inhibition of lipolysis during acute GH exposure increases insulin sensitivity in previously untreated GH-deficient adults. Eur J Endocrinol. 2003;149(6):511–9. doi: 10.1530/eje.0.1490511. [DOI] [PubMed] [Google Scholar]
  • 101.Hansen BC, Jen KC, Belbez Pek S, Wolfe RA. Rapid oscillations in plasma insulin, glucagon, and glucose in obese and normal weight humans. J Clin Endocrinol Metab. 1982;54(4):785–92. doi: 10.1210/jcem-54-4-785. [DOI] [PubMed] [Google Scholar]
  • 102.Lang DA, Matthews DR, Peto J, Turner RC. Cyclic oscillations of basal plasma glucose and insulin concentrations in human beings. N Engl J Med. 1979;301(19):1023–7. doi: 10.1056/NEJM197911083011903. [DOI] [PubMed] [Google Scholar]
  • 103.Koopmans SJ, Sips HC, Krans HM, Radder JK. Pulsatile intravenous insulin replacement in streptozotocin diabetic rats is more efficient than continuous delivery: effects on glycaemic control, insulin-mediated glucose metabolism and lipolysis. Diabetologia. 1996;39(4):391–400. doi: 10.1007/BF00400670. [DOI] [PubMed] [Google Scholar]
  • 104.Matthews DR, Naylor BA, Jones RG, Ward GM, Turner RC. Pulsatile insulin has greater hypoglycemic effect than continuous delivery. Diabetes. 1983;32(7):617–21. doi: 10.2337/diab.32.7.617. [DOI] [PubMed] [Google Scholar]
  • 105.Boucher J, Kleinridders A, Kahn CR. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol. 2014;6(1). doi: 10.1101/cshperspect.a009191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat Med. 2017;23(7):804–14. doi: 10.1038/nm.4350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev. 2018;98(4):2133–223. doi: 10.1152/physrev.00063.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y, Roth J. Insulin resistance and hyperinsulinemia: is hyperinsulinemia the cart or the horse? Diabetes Care. 2008;31 Suppl 2:S262–8. doi: 10.2337/dc08-s264. [DOI] [PubMed] [Google Scholar]
  • 109.Marshall S, Olefsky JM. Effects of insulin incubation on insulin binding, glucose transport, and insulin degradation by isolated rat adipocytes. Evidence for hormone-induced desensitization at the receptor and postreceptor level. J Clin Invest. 1980;66(4):763–72. doi: 10.1172/JCI109914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Catalano KJ, Maddux BA, Szary J, Youngren JF, Goldfine ID, Schaufele F. Insulin resistance induced by hyperinsulinemia coincides with a persistent alteration at the insulin receptor tyrosine kinase domain. PLoS One. 2014;9(9):e108693. doi: 10.1371/journal.pone.0108693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lerario AC, el-Andere W, Wajchenberg BL, Ohnuma LY, Rocha MH, Andriolo A. Insulin resistance in acromegaly: evaluation by studies of insulin binding to erythrocytes. J Endocrinol Invest. 1989;12(3):155–61. doi: 10.1007/BF03349950. [DOI] [PubMed] [Google Scholar]
  • 112.Pav J, Hilgertova J, Marek J, Sramkova J. Insulin binding to erythrocyte receptors in acromegalic patients in relation to the activity of acromegaly and to concomitant diabetes mellitus. Exp Clin Endocrinol. 1986;88(1):76–80. doi: 10.1055/s-0029-1210578. [DOI] [PubMed] [Google Scholar]
  • 113.Muggeo M, Saviolakis GA, Businaro V, Valerio A, Moghetti P, Crepaldi G. Insulin receptor on monocytes from patients with acromegaly and fasting hyperglycemia. J Clin Endocrinol Metab. 1983;56(4):733–8. doi: 10.1210/jcem-56-4-733. [DOI] [PubMed] [Google Scholar]
  • 114.Mavalli MD, DiGirolamo DJ, Fan Y, Riddle RC, Campbell KS, van Groen T, Frank SJ, Sperling MA, Esser KA, Bamman MM, Clemens TL. Distinct growth hormone receptor signaling modes regulate skeletal muscle development and insulin sensitivity in mice. J Clin Invest. 2010;120(11):4007–20. doi: 10.1172/JCI42447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.List EO, Berryman DE, Ikeno Y, Hubbard GB, Funk K, Comisford R, Young JA, Stout MB, Tchkonia T, Masternak MM, Bartke A, Kirkland JL, Miller RA, Kopchick JJ. Removal of growth hormone receptor (GHR) in muscle of male mice replicates some of the health benefits seen in global GHR−/− mice. Aging (Albany NY). 2015;7(7):500–12. doi: 10.18632/aging.100766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vijayakumar A, Wu Y, Sun H, Li X, Jeddy Z, Liu C, Schwartz GJ, Yakar S, LeRoith D. Targeted loss of GHR signaling in mouse skeletal muscle protects against high-fat diet-induced metabolic deterioration. Diabetes. 2012;61(1):94–103. doi: 10.2337/db11-0814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Smith TR, Elmendorf JS, David TS, Turinsky J. Growth hormone-induced insulin resistance: role of the insulin receptor, IRS-1, GLUT-1, and GLUT-4. Am J Physiol. 1997;272(6 Pt 1):E1071–9. doi: 10.1152/ajpendo.1997.272.6.E1071. [DOI] [PubMed] [Google Scholar]
  • 118.Thirone AC, Carvalho CR, Brenelli SL, Velloso LA, Saad MJ. Effect of chronic growth hormone treatment on insulin signal transduction in rat tissues. Mol Cell Endocrinol. 1997;130(1–2):33–42. doi: 10.1016/s0303-7207(97)00071-3. [DOI] [PubMed] [Google Scholar]
  • 119.Barbour LA, Shao J, Qiao L, Leitner W, Anderson M, Friedman JE, Draznin B. Human placental growth hormone increases expression of the p85 regulatory unit of phosphatidylinositol 3-kinase and triggers severe insulin resistance in skeletal muscle. Endocrinology. 2004;145(3):1144–50. doi: 10.1210/en.2003-1297. [DOI] [PubMed] [Google Scholar]
  • 120.Bramnert M, Segerlantz M, Laurila E, Daugaard JR, Manhem P, Groop L. Growth hormone replacement therapy induces insulin resistance by activating the glucose-fatty acid cycle. J Clin Endocrinol Metab. 2003;88(4):1455–63. doi: 10.1210/jc.2002-020542. [DOI] [PubMed] [Google Scholar]
  • 121.Alford FP, Hew FL, Christopher MC, Rantzau C. Insulin sensitivity in growth hormone (GH)-deficient adults and effect of GH replacement therapy. J Endocrinol Invest. 1999;22(5 Suppl):28–32. [PubMed] [Google Scholar]
  • 122.Galimov A, Hartung A, Trepp R, Mader A, Fluck M, Linke A, Bluher M, Christ E, Krutzfeldt J. Growth hormone replacement therapy regulates microRNA-29a and targets involved in insulin resistance. J Mol Med (Berl). 2015;93(12):1369–79. doi: 10.1007/s00109-015-1322-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jessen N, Djurhuus CB, Jorgensen JO, Jensen LS, Moller N, Lund S, Schmitz O. Evidence against a role for insulin-signaling proteins PI 3-kinase and Akt in insulin resistance in human skeletal muscle induced by short-term GH infusion. Am J Physiol Endocrinol Metab. 2005;288(1):E194–9. doi: 10.1152/ajpendo.00149.2004. [DOI] [PubMed] [Google Scholar]
  • 124.Krusenstjerna-Hafstrom T, Clasen BF, Moller N, Jessen N, Pedersen SB, Christiansen JS, Jorgensen JO. Growth hormone (GH)-induced insulin resistance is rapidly reversible: an experimental study in GH-deficient adults. J Clin Endocrinol Metab. 2011;96(8):2548–57. doi: 10.1210/jc.2011-0273. [DOI] [PubMed] [Google Scholar]
  • 125.Nielsen S, Moller N, Christiansen JS, Jorgensen JO. Pharmacological antilipolysis restores insulin sensitivity during growth hormone exposure. Diabetes. 2001;50(10):2301–8. doi: 10.2337/diabetes.50.10.2301. [DOI] [PubMed] [Google Scholar]
  • 126.Segerlantz M, Bramnert M, Manhem P, Laurila E, Groop LC. Inhibition of the rise in FFA by Acipimox partially prevents GH-induced insulin resistance in GH-deficient adults. J Clin Endocrinol Metab. 2001;86(12):5813–8. doi: 10.1210/jcem.86.12.8096. [DOI] [PubMed] [Google Scholar]
  • 127.Forrest L, Sedmak C, Sikder S, Grewal S, Harman SM, Blackman MR, Muniyappa R. Effects of growth hormone on hepatic insulin sensitivity and glucose effectiveness in healthy older adults. Endocrine. 2019;63(3):497–506. doi: 10.1007/s12020-018-01834-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hogild ML, Bak AM, Pedersen SB, Rungby J, Frystyk J, Moller N, Jessen N, Jorgensen JOL. Growth hormone signaling and action in obese versus lean human subjects. Am J Physiol Endocrinol Metab. 2019;316(2):E333–E44. doi: 10.1152/ajpendo.00431.2018. [DOI] [PubMed] [Google Scholar]
  • 129.Nellemann B, Vendelbo MH, Nielsen TS, Bak AM, Hogild M, Pedersen SB, Bienso RS, Pilegaard H, Moller N, Jessen N, Jorgensen JO. Growth hormone-induced insulin resistance in human subjects involves reduced pyruvate dehydrogenase activity. Acta Physiol (Oxf). 2014;210(2):392–402. doi: 10.1111/apha.12183. [DOI] [PubMed] [Google Scholar]
  • 130.Shulman GI. Cellular mechanisms of insulin resistance. J Clin Invest. 2000;106(2):171–6. doi: 10.1172/JCI10583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Perry RJ, Camporez JG, Kursawe R, Titchenell PM, Zhang D, Perry CJ, Jurczak MJ, Abudukadier A, Han MS, Zhang XM, Ruan HB, Yang X, Caprio S, Kaech SM, Sul HS, Birnbaum MJ, Davis RJ, Cline GW, Petersen KF, Shulman GI. Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes. Cell. 2015;160(4):745–58. doi: 10.1016/j.cell.2015.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Moller N, Butler PC, Antsiferov MA, Alberti KG. Effects of growth hormone on insulin sensitivity and forearm metabolism in normal man. Diabetologia. 1989;32(2):105–10. doi: 10.1007/BF00505182. [DOI] [PubMed] [Google Scholar]
  • 133.Orskov L, Schmitz O, Jorgensen JO, Arnfred J, Abildgaard N, Christiansen JS, Alberti KG, Orskov H. Influence of growth hormone on glucose-induced glucose uptake in normal men as assessed by the hyperglycemic clamp technique. J Clin Endocrinol Metab. 1989;68(2):276–82. doi: 10.1210/jcem-68-2-276. [DOI] [PubMed] [Google Scholar]
  • 134.Liu Z, Cordoba-Chacon J, Kineman RD, Cronstein BN, Muzumdar R, Gong Z, Werner H, Yakar S. Growth Hormone Control of Hepatic Lipid Metabolism. Diabetes. 2016;65(12):3598–609. doi: 10.2337/db16-0649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Cordoba-Chacon J, Majumdar N, List EO, Diaz-Ruiz A, Frank SJ, Manzano A, Bartrons R, Puchowicz M, Kopchick JJ, Kineman RD. Growth Hormone Inhibits Hepatic De Novo Lipogenesis in Adult Mice. Diabetes. 2015;64(9):3093–103. doi: 10.2337/db15-0370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol. 2008;9(5):367–77. doi: 10.1038/nrm2391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gao Y, Su P, Wang C, Zhu K, Chen X, Liu S, He J. The role of PTEN in chronic growth hormone-induced hepatic insulin resistance. PLoS One. 2013;8(6):e68105. doi: 10.1371/journal.pone.0068105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Boparai RK, Arum O, Khardori R, Bartke A. Glucose homeostasis and insulin sensitivity in growth hormone-transgenic mice: a cross-sectional analysis. Biol Chem. 2010;391(10):1149–55. doi: 10.1515/BC.2010.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hjortebjerg R, Berryman DE, Comisford R, Frank SJ, List EO, Bjerre M, Frystyk J, Kopchick JJ. Insulin, IGF-1, and GH Receptors Are Altered in an Adipose Tissue Depot-Specific Manner in Male Mice With Modified GH Action. Endocrinology. 2017;158(5):1406–18. doi: 10.1210/en.2017-00084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Venkatesan N, Davidson MB. Insulin resistance in rats harboring growth hormone-secreting tumors: decreased receptor number but increased kinase activity in liver. Metabolism. 1995;44(1):75–84. doi: 10.1016/0026-0495(95)90292-9. [DOI] [PubMed] [Google Scholar]
  • 141.Diaz ME, Gonzalez L, Miquet JG, Martinez CS, Sotelo AI, Bartke A, Turyn D. Growth hormone modulation of EGF-induced PI3K-Akt pathway in mice liver. Cell Signal. 2012;24(2):514–23. doi: 10.1016/j.cellsig.2011.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Dominici FP, Arostegui Diaz G, Bartke A, Kopchick JJ, Turyn D. Compensatory alterations of insulin signal transduction in liver of growth hormone receptor knockout mice. J Endocrinol. 2000;166(3):579–90. doi: 10.1677/joe.0.1660579. [DOI] [PubMed] [Google Scholar]
  • 143.Dominici FP, Hauck S, Argentino DP, Bartke A, Turyn D. Increased insulin sensitivity and upregulation of insulin receptor, insulin receptor substrate (IRS)-1 and IRS-2 in liver of Ames dwarf mice. J Endocrinol. 2002;173(1):81–94. doi: 10.1677/joe.0.1730081. [DOI] [PubMed] [Google Scholar]
  • 144.Bennis MT, Schneider A, Victoria B, Do A, Wiesenborn DS, Spinel L, Gesing A, Kopchick JJ, Siddiqi SA, Masternak MM. The role of transplanted visceral fat from the long-lived growth hormone receptor knockout mice on insulin signaling. Geroscience. 2017;39(1):51–9. doi: 10.1007/s11357-017-9957-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Higham CE, Rowles S, Russell-Jones D, Umpleby AM, Trainer PJ. Pegvisomant improves insulin sensitivity and reduces overnight free fatty acid concentrations in patients with acromegaly. J Clin Endocrinol Metab. 2009;94(7):2459–63. doi: 10.1210/jc.2008-2086. [DOI] [PubMed] [Google Scholar]
  • 146.Pedersen MH, Svart MV, Lebeck J, Bidlingmaier M, Stodkilde-Jorgensen H, Pedersen SB, Moller N, Jessen N, Jorgensen JOL. Substrate Metabolism and Insulin Sensitivity During Fasting in Obese Human Subjects: Impact of GH Blockade. J Clin Endocrinol Metab. 2017;102(4):1340–9. doi: 10.1210/jc.2016-3835. [DOI] [PubMed] [Google Scholar]
  • 147.Corbit KC, Camporez JPG, Edmunds LR, Tran JL, Vera NB, Erion DM, Deo RC, Perry RJ, Shulman GI, Jurczak MJ, Weiss EJ. Adipocyte JAK2 Regulates Hepatic Insulin Sensitivity Independently of Body Composition, Liver Lipid Content, and Hepatic Insulin Signaling. Diabetes. 2018;67(2):208–21. doi: 10.2337/db17-0524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Corbit KC, Camporez JPG, Tran JL, Wilson CG, Lowe DA, Nordstrom SM, Ganeshan K, Perry RJ, Shulman GI, Jurczak MJ, Weiss EJ. Adipocyte JAK2 mediates growth hormone-induced hepatic insulin resistance. JCI Insight. 2017;2(3):e91001. doi: 10.1172/jci.insight.91001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Tran TT, Yamamoto Y, Gesta S, Kahn CR. Beneficial effects of subcutaneous fat transplantation on metabolism. Cell Metab. 2008;7(5):410–20. doi: 10.1016/j.cmet.2008.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.List EO, Berryman DE, Buchman M, Jensen EA, Funk K, Duran-Ortiz S, Qian Y, Young JA, Slyby J, McKenna S, Kopchick JJ. GH Knockout Mice Have Increased Subcutaneous Adipose Tissue With Decreased Fibrosis and Enhanced Insulin Sensitivity. Endocrinology. 2019;160(7):1743–56. doi: 10.1210/en.2019-00167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol. 2017;7(3):819–40. doi: 10.1002/cphy.c160027. [DOI] [PubMed] [Google Scholar]
  • 152.Gjedsted J, Gormsen LC, Nielsen S, Schmitz O, Djurhuus CB, Keiding S, Orskov H, Tonnesen E, Moller N. Effects of a 3-day fast on regional lipid and glucose metabolism in human skeletal muscle and adipose tissue. Acta Physiol (Oxf). 2007;191(3):205–16. doi: 10.1111/j.1748-1716.2007.01740.x. [DOI] [PubMed] [Google Scholar]
  • 153.Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond). 2010;34(6):949–59. doi: 10.1038/ijo.2009.286. [DOI] [PubMed] [Google Scholar]
  • 154.Nielsen TS, Jessen N, Jorgensen JO, Moller N, Lund S. Dissecting adipose tissue lipolysis: molecular regulation and implications for metabolic disease. J Mol Endocrinol. 2014;52(3):R199–222. doi: 10.1530/JME-13-0277. [DOI] [PubMed] [Google Scholar]
  • 155.Yang S, Mulder H, Holm C, Eden S. Effects of growth hormone on the function of beta-adrenoceptor subtypes in rat adipocytes. Obes Res. 2004;12(2):330–9. doi: 10.1038/oby.2004.41. [DOI] [PubMed] [Google Scholar]
  • 156.Giralt M, Villarroya F. White, brown, beige/brite: different adipose cells for different functions? Endocrinology. 2013;154(9):2992–3000. doi: 10.1210/en.2013-1403. [DOI] [PubMed] [Google Scholar]
  • 157.Lee J, Ellis JM, Wolfgang MJ. Adipose fatty acid oxidation is required for thermogenesis and potentiates oxidative stress-induced inflammation. Cell Rep. 2015;10(2):266–79. doi: 10.1016/j.celrep.2014.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Lee JH, Park A, Oh KJ, Lee SC, Kim WK, Bae KH. The Role of Adipose Tissue Mitochondria: Regulation of Mitochondrial Function for the Treatment of Metabolic Diseases. Int J Mol Sci. 2019;20(19). doi: 10.3390/ijms20194924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Park A, Kim WK, Bae KH. Distinction of white, beige and brown adipocytes derived from mesenchymal stem cells. World J Stem Cells. 2014;6(1):33–42. doi: 10.4252/wjsc.v6.i1.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Shabalina IG, Petrovic N, de Jong JM, Kalinovich AV, Cannon B, Nedergaard J. UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic. Cell Rep. 2013;5(5):1196–203. doi: 10.1016/j.celrep.2013.10.044. [DOI] [PubMed] [Google Scholar]
  • 161.Jager S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci U S A. 2007;104(29):12017–22. doi: 10.1073/pnas.0705070104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.De Matteis R, Lucertini F, Guescini M, Polidori E, Zeppa S, Stocchi V, Cinti S, Cuppini R. Exercise as a new physiological stimulus for brown adipose tissue activity. Nutr Metab Cardiovasc Dis. 2013;23(6):582–90. doi: 10.1016/j.numecd.2012.01.013. [DOI] [PubMed] [Google Scholar]
  • 163.Nelson CN, List EO, Ieremia M, Constantin L, Chhabra Y, Kopchick JJ, Waters MJ. Growth hormone activated STAT5 is required for induction of beige fat in vivo. Growth Horm IGF Res. 2018;42–43:40–51. doi: 10.1016/j.ghir.2018.08.004. [DOI] [PubMed] [Google Scholar]
  • 164.Moran A, Jacobs DR Jr., Steinberger J, Cohen P, Hong CP, Prineas R, Sinaiko AR. Association between the insulin resistance of puberty and the insulin-like growth factor-I/growth hormone axis. J Clin Endocrinol Metab. 2002;87(10):4817–20. doi: 10.1210/jc.2002-020517. [DOI] [PubMed] [Google Scholar]
  • 165.Bloch CA, Clemons P, Sperling MA. Puberty decreases insulin sensitivity. J Pediatr. 1987;110(3):481–7. doi: 10.1016/s0022-3476(87)80522-x. [DOI] [PubMed] [Google Scholar]
  • 166.Caprio S, Cline G, Boulware S, Permanente C, Shulman GI, Sherwin RS, Tamborlane WV. Effects of puberty and diabetes on metabolism of insulin-sensitive fuels. Am J Physiol. 1994;266(6 Pt 1):E885–91. doi: 10.1152/ajpendo.1994.266.6.E885. [DOI] [PubMed] [Google Scholar]
  • 167.Goran MI, Gower BA. Longitudinal study on pubertal insulin resistance. Diabetes. 2001;50(11):2444–50. doi: 10.2337/diabetes.50.11.2444. [DOI] [PubMed] [Google Scholar]
  • 168.Moran A, Jacobs DR Jr., Steinberger J, Hong CP, Prineas R, Luepker R, Sinaiko AR. Insulin resistance during puberty: results from clamp studies in 357 children. Diabetes. 1999;48(10):2039–44. doi: 10.2337/diabetes.48.10.2039. [DOI] [PubMed] [Google Scholar]
  • 169.Ahn CW, Kim CS, Nam JH, Kim HJ, Nam JS, Park JS, Kang ES, Cha BS, Lim SK, Kim KR, Lee HC, Huh KB. Effects of growth hormone on insulin resistance and atherosclerotic risk factors in obese type 2 diabetic patients with poor glycaemic control. Clin Endocrinol (Oxf). 2006;64(4):444–9. doi: 10.1111/j.1365-2265.2006.02490.x. [DOI] [PubMed] [Google Scholar]
  • 170.Nam SY, Kim KR, Cha BS, Song YD, Lim SK, Lee HC, Huh KB. Low-dose growth hormone treatment combined with diet restriction decreases insulin resistance by reducing visceral fat and increasing muscle mass in obese type 2 diabetic patients. Int J Obes Relat Metab Disord. 2001;25(8):1101–7. doi: 10.1038/sj.ijo.0801636. [DOI] [PubMed] [Google Scholar]
  • 171.Nellemann B, Gormsen LC, Sorensen LP, Christiansen JS, Nielsen S. Impaired insulin-mediated antilipolysis and lactate release in adipose tissue of upper-body obese women. Obesity (Silver Spring). 2012;20(1):57–64. doi: 10.1038/oby.2011.290. [DOI] [PubMed] [Google Scholar]
  • 172.Rizza RA, Mandarino LJ, Gerich JE. Effects of growth hormone on insulin action in man. Mechanisms of insulin resistance, impaired suppression of glucose production, and impaired stimulation of glucose utilization. Diabetes. 1982;31(8 Pt 1):663–9. doi: 10.2337/diab.31.8.663. [DOI] [PubMed] [Google Scholar]
  • 173.Vijayakumar A, Yakar S, Leroith D. The intricate role of growth hormone in metabolism. Front Endocrinol (Lausanne). 2011;2:32. doi: 10.3389/fendo.2011.00032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Bengtsson BA, Brummer RJ, Eden S, Bosaeus I, Lindstedt G. Body composition in acromegaly: the effect of treatment. Clin Endocrinol (Oxf). 1989;31(4):481–90. doi: 10.1111/j.1365-2265.1989.tb01272.x. [DOI] [PubMed] [Google Scholar]
  • 175.Mercado M, Ramirez-Renteria C. Metabolic Complications of Acromegaly. Front Horm Res. 2018;49:20–8. doi: 10.1159/000486001. [DOI] [PubMed] [Google Scholar]
  • 176.Olarescu NC, Bollerslev J. The Impact of Adipose Tissue on Insulin Resistance in Acromegaly. Trends Endocrinol Metab. 2016;27(4):226–37. doi: 10.1016/j.tem.2016.02.005. [DOI] [PubMed] [Google Scholar]
  • 177.Unal A, Sahin Y, Kelestimur F. Acromegaly with polycystic ovaries, hyperandrogenism, hirsutism, insulin resistance and acanthosis nigricans: a case report. Endocr J. 1993;40(2):207–11. doi: 10.1507/endocrj.40.207. [DOI] [PubMed] [Google Scholar]
  • 178.Berryman DE, List EO, Coschigano KT, Behar K, Kim JK, Kopchick JJ. Comparing adiposity profiles in three mouse models with altered GH signaling. Growth Horm IGF Res. 2004;14(4):309–18. doi: 10.1016/j.ghir.2004.02.005. [DOI] [PubMed] [Google Scholar]
  • 179.Coates RJ, Kolor K, Stewart SL, Richardson LC. Diagnostic markers for ovarian cancer screening: not ready for routine clinical use. Clin Cancer Res. 2008;14(22):7575–6; author reply 7–9. doi: 10.1158/1078-0432.CCR-08-2296. [DOI] [PubMed] [Google Scholar]
  • 180.Olsson B, Bohlooly YM, Fitzgerald SM, Frick F, Ljungberg A, Ahren B, Tornell J, Bergstrom G, Oscarsson J. Bovine growth hormone transgenic mice are resistant to diet-induced obesity but develop hyperphagia, dyslipidemia, and diabetes on a high-fat diet. Endocrinology. 2005;146(2):920–30. doi: 10.1210/en.2004-1232. [DOI] [PubMed] [Google Scholar]
  • 181.Rosenbaum M, Gertner JM, Leibel RL. Effects of systemic growth hormone (GH) administration on regional adipose tissue distribution and metabolism in GH-deficient children. J Clin Endocrinol Metab. 1989;69(6):1274–81. doi: 10.1210/jcem-69-6-1274. [DOI] [PubMed] [Google Scholar]
  • 182.Laron Z, Ginsberg S, Lilos P, Arbiv M, Vaisman N. Body composition in untreated adult patients with Laron syndrome (primary GH insensitivity). Clin Endocrinol (Oxf). 2006;65(1):114–7. doi: 10.1111/j.1365-2265.2006.02558.x. [DOI] [PubMed] [Google Scholar]
  • 183.Hill CM, Fang Y, Miquet JG, Sun LY, Masternak MM, Bartke A. Long-lived hypopituitary Ames dwarf mice are resistant to the detrimental effects of high-fat diet on metabolic function and energy expenditure. Aging Cell. 2016;15(3):509–21. doi: 10.1111/acel.12467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Moller L, Dalman L, Norrelund H, Billestrup N, Frystyk J, Moller N, Jorgensen JO. Impact of fasting on growth hormone signaling and action in muscle and fat. J Clin Endocrinol Metab. 2009;94(3):965–72. doi: 10.1210/jc.2008-1385. [DOI] [PubMed] [Google Scholar]
  • 185.Norrelund H, Moller N, Nair KS, Christiansen JS, Jorgensen JO. Continuation of growth hormone (GH) substitution during fasting in GH-deficient patients decreases urea excretion and conserves protein synthesis. J Clin Endocrinol Metab. 2001;86(7):3120–9. doi: 10.1210/jcem.86.7.7618. [DOI] [PubMed] [Google Scholar]
  • 186.Norrelund H, Nair KS, Nielsen S, Frystyk J, Ivarsen P, Jorgensen JO, Christiansen JS, Moller N. The decisive role of free fatty acids for protein conservation during fasting in humans with and without growth hormone. J Clin Endocrinol Metab. 2003;88(9):4371–8. doi: 10.1210/jc.2003-030267. [DOI] [PubMed] [Google Scholar]
  • 187.Vendelbo MH, Jorgensen JO, Pedersen SB, Gormsen LC, Lund S, Schmitz O, Jessen N, Moller N. Exercise and fasting activate growth hormone-dependent myocellular signal transducer and activator of transcription-5b phosphorylation and insulin-like growth factor-I messenger ribonucleic acid expression in humans. J Clin Endocrinol Metab. 2010;95(9):E64–8. doi: 10.1210/jc.2010-0689. [DOI] [PubMed] [Google Scholar]
  • 188.Jorgensen JO, Moller N, Lauritzen T, Alberti KG, Orskov H, Christiansen JS. Evening versus morning injections of growth hormone (GH) in GH-deficient patients: effects on 24-hour patterns of circulating hormones and metabolites. J Clin Endocrinol Metab. 1990;70(1):207–14. doi: 10.1210/jcem-70-1-207. [DOI] [PubMed] [Google Scholar]
  • 189.Collins S beta-Adrenoceptor Signaling Networks in Adipocytes for Recruiting Stored Fat and Energy Expenditure. Front Endocrinol (Lausanne). 2011;2:102. doi: 10.3389/fendo.2011.00102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hjelholt AJ, Lee KY, Arlien-Soborg MC, Pedersen SB, Kopchick JJ, Puri V, Jessen N, Jorgensen JOL. Temporal patterns of lipolytic regulators in adipose tissue after acute growth hormone exposure in human subjects: A randomized controlled crossover trial. Mol Metab. 2019;29:65–75. doi: 10.1016/j.molmet.2019.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Nielsen TS, Vendelbo MH, Jessen N, Pedersen SB, Jorgensen JO, Lund S, Moller N. Fasting, but not exercise, increases adipose triglyceride lipase (ATGL) protein and reduces G(0)/G(1) switch gene 2 (G0S2) protein and mRNA content in human adipose tissue. J Clin Endocrinol Metab. 2011;96(8):E1293–7. doi: 10.1210/jc.2011-0149. [DOI] [PubMed] [Google Scholar]
  • 192.Hansen TK, Gravholt CH, H OR, Rasmussen MH, Christiansen JS, Jorgensen JO. Dose dependency of the pharmacokinetics and acute lipolytic actions of growth hormone. J Clin Endocrinol Metab. 2002;87(10):4691–8. doi: 10.1210/jc.2002-020563. [DOI] [PubMed] [Google Scholar]
  • 193.Richelsen B Action of growth hormone in adipose tissue. Horm Res. 1997;48 Suppl 5:105–10. doi: 10.1159/000191338. [DOI] [PubMed] [Google Scholar]
  • 194.del Rincon JP, Iida K, Gaylinn BD, McCurdy CE, Leitner JW, Barbour LA, Kopchick JJ, Friedman JE, Draznin B, Thorner MO. Growth hormone regulation of p85alpha expression and phosphoinositide 3-kinase activity in adipose tissue: mechanism for growth hormone-mediated insulin resistance. Diabetes. 2007;56(6):1638–46. doi: 10.2337/db06-0299. [DOI] [PubMed] [Google Scholar]
  • 195.Fain JN, Cheema P, Tichansky DS, Madan AK. Stimulation of human omental adipose tissue lipolysis by growth hormone plus dexamethasone. Mol Cell Endocrinol. 2008;295(1–2):101–5. doi: 10.1016/j.mce.2008.05.014. [DOI] [PubMed] [Google Scholar]
  • 196.Johansen T, Richelsen B, Hansen HS, Din N, Malmlof K. Growth hormone-mediated breakdown of body fat: effects of GH on lipases in adipose tissue and skeletal muscle of old rats fed different diets. Horm Metab Res. 2003;35(4):243–50. doi: 10.1055/s-2003-39481. [DOI] [PubMed] [Google Scholar]
  • 197.Ng FM, Jiang WJ, Gianello R, Pitt S, Roupas P. Molecular and cellular actions of a structural domain of human growth hormone (AOD9401) on lipid metabolism in Zucker fatty rats. J Mol Endocrinol. 2000;25(3):287–98. doi: 10.1677/jme.0.0250287. [DOI] [PubMed] [Google Scholar]
  • 198.Richelsen B, Pedersen SB, Kristensen K, Borglum JD, Norrelund H, Christiansen JS, Jorgensen JO. Regulation of lipoprotein lipase and hormone-sensitive lipase activity and gene expression in adipose and muscle tissue by growth hormone treatment during weight loss in obese patients. Metabolism. 2000;49(7):906–11. doi: 10.1053/meta.2000.6738. [DOI] [PubMed] [Google Scholar]
  • 199.Brasaemle DL, Dolios G, Shapiro L, Wang R. Proteomic analysis of proteins associated with lipid droplets of basal and lipolytically stimulated 3T3-L1 adipocytes. J Biol Chem. 2004;279(45):46835–42. doi: 10.1074/jbc.M409340200. [DOI] [PubMed] [Google Scholar]
  • 200.Kershaw EE, Schupp M, Guan HP, Gardner NP, Lazar MA, Flier JS. PPARgamma regulates adipose triglyceride lipase in adipocytes in vitro and in vivo. Am J Physiol Endocrinol Metab. 2007;293(6):E1736–45. doi: 10.1152/ajpendo.00122.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Kim YJ, Cho SY, Yun CH, Moon YS, Lee TR, Kim SH. Transcriptional activation of Cidec by PPARgamma2 in adipocyte. Biochem Biophys Res Commun. 2008;377(1):297–302. doi: 10.1016/j.bbrc.2008.09.129. [DOI] [PubMed] [Google Scholar]
  • 202.Zandbergen F, Mandard S, Escher P, Tan NS, Patsouris D, Jatkoe T, Rojas-Caro S, Madore S, Wahli W, Tafuri S, Muller M, Kersten S. The G0/G1 switch gene 2 is a novel PPAR target gene. Biochem J. 2005;392(Pt 2):313–24. doi: 10.1042/BJ20050636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.de la Rosa Rodriguez MA, Kersten S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862(10 Pt B):1212–20. doi: 10.1016/j.bbalip.2017.07.007. [DOI] [PubMed] [Google Scholar]
  • 204.Varga T, Czimmerer Z, Nagy L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim Biophys Acta. 2011;1812(8):1007–22. doi: 10.1016/j.bbadis.2011.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Georgiadi A, Kersten S. Mechanisms of gene regulation by fatty acids. Adv Nutr. 2012;3(2):127–34. doi: 10.3945/an.111.001602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Grahn TH, Kaur R, Yin J, Schweiger M, Sharma VM, Lee MJ, Ido Y, Smas CM, Zechner R, Lass A, Puri V. Fat-specific protein 27 (FSP27) interacts with adipose triglyceride lipase (ATGL) to regulate lipolysis and insulin sensitivity in human adipocytes. J Biol Chem. 2014;289(17):12029–39. doi: 10.1074/jbc.M113.539890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Grahn TH, Zhang Y, Lee MJ, Sommer AG, Mostoslavsky G, Fried SK, Greenberg AS, Puri V. FSP27 and PLIN1 interaction promotes the formation of large lipid droplets in human adipocytes. Biochem Biophys Res Commun. 2013;432(2):296–301. doi: 10.1016/j.bbrc.2013.01.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Jambunathan S, Yin J, Khan W, Tamori Y, Puri V. FSP27 promotes lipid droplet clustering and then fusion to regulate triglyceride accumulation. PLoS One. 2011;6(12):e28614. doi: 10.1371/journal.pone.0028614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Keller P, Petrie JT, De Rose P, Gerin I, Wright WS, Chiang SH, Nielsen AR, Fischer CP, Pedersen BK, MacDougald OA. Fat-specific protein 27 regulates storage of triacylglycerol. J Biol Chem. 2008;283(21):14355–65. doi: 10.1074/jbc.M708323200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Puri V, Konda S, Ranjit S, Aouadi M, Chawla A, Chouinard M, Chakladar A, Czech MP. Fat-specific protein 27, a novel lipid droplet protein that enhances triglyceride storage. J Biol Chem. 2007;282(47):34213–8. doi: 10.1074/jbc.M707404200. [DOI] [PubMed] [Google Scholar]
  • 211.Puri V, Ranjit S, Konda S, Nicoloro SM, Straubhaar J, Chawla A, Chouinard M, Lin C, Burkart A, Corvera S, Perugini RA, Czech MP. Cidea is associated with lipid droplets and insulin sensitivity in humans. Proc Natl Acad Sci U S A. 2008;105(22):7833–8. doi: 10.1073/pnas.0802063105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Slayton M, Gupta A, Balakrishnan B, Puri V. CIDE Proteins in Human Health and Disease. Cells. 2019;8(3). doi: 10.3390/cells8030238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Singh M, Kaur R, Lee MJ, Pickering RT, Sharma VM, Puri V, Kandror KV. Fat-specific protein 27 inhibits lipolysis by facilitating the inhibitory effect of transcription factor Egr1 on transcription of adipose triglyceride lipase. J Biol Chem. 2014;289(21):14481–7. doi: 10.1074/jbc.C114.563080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Bergan-Roller HE, Ickstadt AT, Kittilson JD, Sheridan MA. Insulin and insulin-like growth factor-1 modulate the lipolytic action of growth hormone by altering signal pathway linkages. Gen Comp Endocrinol. 2017;248:40–8. doi: 10.1016/j.ygcen.2017.04.005. [DOI] [PubMed] [Google Scholar]
  • 215.Bergan-Roller HE, Sheridan MA. The growth hormone signaling system: Insights into coordinating the anabolic and catabolic actions of growth hormone. Gen Comp Endocrinol. 2018;258:119–33. doi: 10.1016/j.ygcen.2017.07.028. [DOI] [PubMed] [Google Scholar]
  • 216.Carter-Su C, Schwartz J, Argetsinger LS. Growth hormone signaling pathways. Growth Horm IGF Res. 2016;28:11–5. doi: 10.1016/j.ghir.2015.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Burgermeister E, Chuderland D, Hanoch T, Meyer M, Liscovitch M, Seger R. Interaction with MEK causes nuclear export and downregulation of peroxisome proliferator-activated receptor gamma. Mol Cell Biol. 2007;27(3):803–17. doi: 10.1128/MCB.00601-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Tan X, Cao Z, Li M, Xu E, Wang J, Xiao Y. TNF-alpha downregulates CIDEC via MEK/ERK pathway in human adipocytes. Obesity (Silver Spring). 2016;24(5):1070–80. doi: 10.1002/oby.21436. [DOI] [PubMed] [Google Scholar]

RESOURCES