Open access peer-reviewed chapter

Growth Hormone Receptor Signaling Pathways and its Negative Regulation by SOCS2

Written By

Leandro Fernández-Pérez, Amilcar Flores-Morales, Borja Guerra, Juan C. Díaz-Chico and Diego Iglesias-Gato

Submitted: 10 November 2015 Reviewed: 14 June 2016 Published: 12 October 2016

DOI: 10.5772/64606

From the Edited Volume

Restricted Growth - Clinical, Genetic and Molecular Aspects

Edited by Maria del Carmen Cardenas- Aguayo

Chapter metrics overview

2,395 Chapter Downloads

View Full Metrics

Abstract

Growth hormone (GH) is a critical regulator of linear body growth during childhood but continues to have important metabolic actions throughout life. The GH receptor (GHR) is ubiquitously expressed, and deficiency of GHR signaling causes a dramatic impact on normal physiology during somatic development, adulthood, and aging. GHR belongs to a family of receptors without intrinsic kinase activity. However, GH binding to homodimers of GHR results in a conformational change in the receptors and the associated tyrosine kinase Janus kinase 2 (JAK2) molecules. Activated JAK2 phosphorylates the GHR cytoplasmic domain on tyrosine residues, and subsequent JAK2-dependent and JAK2-independent intracellular signal transduction pathways evoke cell responses including changes in gene transcription, proliferation, cytoskeletal reorganization, and lipid and glucose metabolism. JAK2 phosphorylates STAT5b, which is a key transcription factor in GH regulation of target genes associated with body growth, intermediate metabolism, and gender dimorphism; although STAT1, 3, and 5a have also been shown to be recruited by the GHR. In addition, many transcripts are regulated independently of STAT5b as a result of GHR activation of Src, ERK, and PI3K-mTOR signaling pathways. The analysis of molecular mechanisms involved in inactivation of GHR-dependent signaling pathway is also imperative for understanding GH physiology. This is clearly illustrated in the case of hepatic GHR-JAK2-STAT5b activation where signal duration regulates gender differences in liver gene expression. An early step in the termination of GH-dependent signaling is removal of GHRs by endocytosis and ubiquitination. The level of ubiquitin ligase SOCS2 is constitutively low, but its expression is rapidly induced by GH. SOCS2 binding to GHR complex promotes their ubiquitination and subsequent proteasomal degradation, contributing to the termination of the GH intracellular signaling. Clinically relevant, SOCS2 is a key negative regulator of GH-dependent body growth and lipid and glucose homeostasis. Furthermore, several cytokines, growth factors, xenobiotics, and sex hormones can regulate SOCS2 protein level, which provides a mechanism for cross-talking where multiple factors can regulate GHR signaling during somatic development. A better understanding of this complex regulation in physiological and pathological states will contribute to prevent health damage and improve clinical management of patients with growth and metabolic disorders.

Keywords

  • GHR
  • SOCS2
  • Growth
  • Metabolism
  • sexual dimorphism

1. Introduction

Growth hormone (GH) is the main regulator of somatic growth through pleiotropic actions on systemic metabolism and local actions on the bone growth plate [14]. GH is predominantly linked to linear growth during childhood but continues to have important metabolic actions throughout life. Secreted from the pituitary gland, GH binds to its receptor (GHR) on the surface of the cells of the target tissues triggering a rapid cascade of intracellular signaling events, which leads to the expression of GH-regulated genes. This set of genes includes positive regulators of GH actions such as the one coding for the growth factor IGF-I, and also genes involved in the negative feedback mechanism responsible for the termination of the GHR intracellular signal, such as the suppressor of cytokine signaling 2 (SOCS2). In this chapter, we will review the current knowledge about the intracellular GH signaling as well as the role of SOCS2 in this regulation and discuss the implications on body growth.

Advertisement

2. Intracellular GH signaling

GH exerts its intracellular actions via the GHR that is ubiquitously expressed (e.g., liver, fat, muscle, bone, and lymphocytes). The GHR belongs to a family of transmembrane cytokine receptors that lack intrinsic enzymatic activity [5]. Instead, in order to activate intracellular signaling, the GHR cytosolic domain associates to the tyrosine kinase Janus kinase 2 (JAK2) [5]. Upon binding, GH promotes the dimerization of two GHR proteins, which results in a conformational change that triggers the activation of the associated tyrosine kinase JAK2 due to the unmasking of their kinase domain [5]. JAK2 activation triggers cross-phosphorylation event on the two adjacent JAK2 proteins and the phosphorylation of tyrosine residues on the cytosolic domain of the GHR. Signal transducer and activator of transcription (STAT) proteins are then recruited to these phosphorylated tyrosines (pY), where they themselves become substrates of JAK2. Although STAT1, STAT3, and STAT5a can also be recruited to the GHR, STAT5b is the main mediator of GH signaling [1, 3, 6]. Phosphorylation by JAK2 releases STAT5b from the receptor and promotes the formation of STAT5b dimer complexes. STAT5b homodimers translocate to the nucleus, bind to their response elements (TTCNNNGAA) on the DNA, and regulate the transcription of GH target genes (e.g., IGF-1, SOCS2, CYP2C12, and HNF6 [710]). Studies of human subjects carrying rare inactivation mutations in the GHR, STAT5b, and IGF-I genes have demonstrated the essentiality of this pathway for normal human growth. Individuals carrying these mutations exhibit severe dwarfisms with very similar growth curves [1115]. Although STAT proteins are critical for many actions of GH [6], GH activation of JAK2 can initiate signaling pathways in addition to the STAT transcription factors as a result of GHR activation of: (1) the MAPK (Mitogen Activated Protein Kinase) pathway; (2) insulin receptor substrate (IRS) proteins implicated in the activation of the phosphatidylinositol-3-kinase (PI3K) and Akt pathway; (3) signal regulatory protein α (SIRPα), a transmembrane scaffold protein that recruits proteins including the tyrosine phosphatase SHP2; and (4) SH2B1, a scaffold protein that can activate JAK2 and enhance GH regulation of the actin cytoskeleton [6].

Advertisement

3. SOCS2 mediates GHR turnover

The analysis of molecular mechanisms involved in the inactivation of the GHR-dependent signaling pathway is also imperative for understanding GH physiology. This is clearly illustrated in the case of hepatic GHR-JAK2-STAT5b activation where signal duration regulates gender differences in liver gene expression [10]. Studies on primary hepatocytes and several cell lines have shown that GH-induced activation of JAK2-STAT5b is transient, with maximal activation achieved within the first 30 min of stimulation, followed by a period of inactivation. This period is characterized by an inability to achieve maximal JAK2-STAT5b activation by GH in the following 3–4 h, unless GH is withdrawn from the media [16]. Similarly, the male pattern of pituitary GH secretion in rats is episodic with peaks every 3–4 h with unmeasurable basal levels. Consequently, intracellular activation of STAT5b is also episodic and periods with low GH circulating levels are required to achieve maximal activation of STAT5b. On the other hand, female rats, which exhibit a more continuous GH secretion pattern with higher basal levels and smaller, irregular, and intermittent peaks, show reduced STAT5b activation compared with their males counterparts [17].

An early and important step in the termination of GH-dependent signaling is the removal of GHRs from the cell surface by mechanisms of endocytosis, which are dependent on ubiquitination [18]. SOCS2 is part of an E3 ubiquitin ligase complex with a key role in the negative regulation of GHR-JAK2-STAT5b signaling pathway, acting in a classical negative feedback loop manner [1922]. The transcription of SOCS2 is induced by STAT5b in response to GH stimulation—which leads to elevated SOCS2 protein levels [16]—consistent with the critical role of STAT5b for growth [7] (Figure 1). SOCS2 binds to phosphorylated tyrosines at the GHR cytosolic domain through its SH2 domain while recruits Elongin B and C, the scaffold protein Cullin5, and the ring finger protein Rbx2 through its SOCS box domain to assemble an E3 ubiquitin ligase complex that ubiquitinates the GHR and promotes its internalization [23, 24] (Figure 2). These GHR-containing early endosomes are later fused to the lysosomes leading to GHR degradation [24]. In addition to SOCS2, the ubiquitin ligase β-TRCP has also been shown to mediate GHR ubiquitination and internalization with the key difference that this process is not GH dependent and the mechanism seems to act independently from SOCS2 [25]. Therefore, the current evidence would suggest that GHR membrane content is controlled by ubiquitin driven endocytosis [18]. The constitutive internalization of GHR is mediated by β-TRCP, which is further enhanced upon GH induction of SOCS2 expression and function. Furthermore, the SH2 domain of SOCS2 can also bind to other components of the signaling cascade interfering with the propagation of the signal. Particularly, SOCS2 binds the GHR at Tyr487 and Tyr595 to prevent GHR signaling [21, 22]. The activation loop of JAK2 is also a target of SOCS2 that prevents JAK2 tyrosine phosphorylation and activation of STATs [16].

Figure 1.

Negative regulation of GHR-STAT5b signaling pathway by SOCS2: The JAK2-STAT5b signal transduction pathway requires an exquisite cellular control and loss of its regulation can promote alterations in body growth. Intracellular activation of GHR-JAK2-STAT5b signaling pathway is transient and it is followed by a period of inactivation that is characterized by an inability to achieve maximal JAK2-STAT5b activation by GH. GHR-STAT5b activity induces the transcription of SOCS2 that acts as part of an E3 ubiquitin ligase complex that plays a key role in the negative regulation of GHR-JAK2-STAT5b signaling pathway.

The physiological role of SOCS2 as negative regulator of GH signaling was clearly demonstrated after the engineering of SOCS2-/- mice that are 40% larger than their wild-type (WT) mates. This phenotype of enlarged growth is not observed in mice lacking other members of the SOCS family such as SOCS1 or CIS (Cytokine-inducible SH2-containing protein), which strengthen the role of SOCS2 as key negative regulator of GH signaling. These in vivo studies highlight the role of SOCS2 as a key negative regulator of GH-dependent signaling and its role in the control of lipid and glucose homeostasis [26]. High SOCS2 expression levels have been found in the liver and the heart [16, 27], and, importantly, SOCS2 actions may not be confined to regulating GH signaling. There is evidence that SOCS2 can directly bind the IGF-IR, and, therefore, it is possible that SOCS2 also regulates IGF-I signaling, although IGF-I does not induce SOCS2 expression [28, 29]. In addition, SOCS2 has been shown to inhibit signaling by IL-6, LIF, IGF-I, and prolactin (Prl) [19].

Figure 2.

E3 ligase activity of SOCS2 and GHR degradation by proteasome: SOCS2 is part of an E3 ubiquitin ligase complex with a key role in the negative regulation of GHR-JAK2-STAT5b signaling pathway. SOCS2 binds to phosphorylated tyrosines (pY) at GHR cytosolic domain while recruits Elongin B and C, the scaffold protein Cullin5, and the ring finger protein Rbx2 to assemble an E3 ubiquitin ligase complex that mediates ubiquitination of target proteins (e.g., GHR) and their subsequent proteasomal degradation. The activation loop of JAK2 is also a target of SOCS2 that prevents JAK2 tyrosine phosphorylation and activation of STATs.

Advertisement

4. Other mechanisms of GHR signaling inhibition

In addition to SOCS2, GHR signaling induces the expression of other SOCS family members (SOCS-1, -3, and CIS) in a transient fashion. In vitro studies shown that SOCS1, SOCS3, and to a lesser extent CIS, are able to inhibit GHR signaling [30, 31]. Both SOCS1 and SOCS3 can directly bind phosphorylated JAK proteins via their SH2 domains inhibiting the kinase activity through the N-terminal kinase inhibitory region (KIR) [32]. Knockouts (-/-) of SOCS1 and SOCS3 genes are incompatible with life. SOCS1-/- mice died soon after birth due to hyperactivation of INFγ signaling. Combination of SOCS1 deletion with genetic depletion of INFγ or treatment with anti-INFγ antibodies ameliorates this phenotype [33]. Under these conditions, no changes in body growth have been reported, suggesting that SOCS1 may not play a major role in GHR signaling inhibition in vivo. On the other hand, CIS-/- mice exhibit no obvious phenotype, although overexpression of CIS results in restricted growth [16, 34]. Despite certain degree of redundancy within the SOCS family, the results obtained from mouse models suggest that SOCS2 is a major negative regulator of GH signaling in vivo. In addition to SOCS, the level of cell surface GHRs can be influenced by transcriptional, translational, and posttranslational factors (e.g., nutritional status, endocrine context, developmental stage, and sex steroids), which thereby regulate cell sensitivity to GH actions [16]. GHR translocation is also directly inhibited by IGF-I, likely contributing to a local feedback loop to hamper GH sensitivity [35]. As mutations in the GHR [15] and STAT5b genes [11] result in growth deficiencies, activating mutations of the tyrosine phosphatase SHP2 (PTPN11) also generates growth retardation linked to the Noonan syndrome [36]. PTPN11 is involved in the negative regulation of GH signaling, rendering lower IGF-I production after GH stimulation through regulation of the RAS/ERK1/2 pathway. Noonan syndrome driving PTPN11 mutants hyperactivates the RAS/ERK pathway in response to GH in vitro and in vivo, suggesting that GH-induced ERK1/2 activation could contribute to GHR signaling inactivation [36]. In the future, it will be interesting to study whether hyperactivation of ERK1/2 inhibit GHR through induction of SOCS expression [37]. Other protein phosphatases such as SHP1 and PTP1b (encoded by the PTPN6 and PTN1 genes, respectively) also play a role in controlling intracellular GH signaling. Upon GH stimulation, SHP1 translocates into the nucleus, where it binds to STAT5b inhibiting its activity [38]. SHP1 also interacts with phosphorylated JAK2 after GH stimulation, inhibiting the propagation of the signal, accordingly, SHP1-/- mice show prolonged GH signaling [39]. On the other hand, PTP1b is able to dephosphorylate GH-activated GHR and JAK2 [16, 40, 41].

Advertisement

5. GH regulation of body growth

Currently, it is accepted that GH is predominantly linked with postnatal growth, whereas IGF-I is linked with both pre and postnatal growth [42, 43]. The original somatomedin hypothesis, which proposed that pituitary GH increases tissue growth by stimulating production of hepatic IGF-I, has developed gradually, from a simple to a more complex form, by studies showing that (1) GH and IGF-I have both dual and overlapping functions on growth plate [44, 45]. However, there are still unanswered questions about the independent and combined relationships of GH and IGF-I on the growth plate and bone growth, including whether or not GH mediates any IGF-I independent effects on bone growth [46] and (2) conditional liver-specific IGF-I null mice exhibited body weights that were indistinguishable from wild-type littermates [47, 48]. These studies showed that, although the liver is the main source of circulating IGF-I, it is local IGF-I that is important for regulating postnatal growth [47]. Indeed, stimulation of hepatic IGF-I production in IGF-I null mice demonstrated that liver derived endocrine IGF-I contributes to 30% of adult body size and sustains postnatal development [49, 50]. In addition, GH is more effective than IGF-I because GH exerts additional growth-promoting actions independent of IGF-I. Previous studies have nicely demonstrated that STAT5b is important for sexual dimorphism of body growth (male-specific body growth) and liver gene expression [51]. Indeed, GH-dependent transcription of IGF-I is directly regulated by STAT5 [3], and the mode of GH administration (i.e., continuous vs. intermittent) influences GH actions on body growth rate, IGF-I expression, and STAT5b activity, which might be clinically relevant. Intermittent (male-like pattern) GH administration to rodents is a more potent stimulus of body growth rate, IGF-I expression, and STAT5b activity in liver than is continuous (female-like pattern) administration [17]. Global disruption of STAT5b in mice caused loss of sexually dimorphic growth characteristics, so that the affected males reduced their size to female size while female mice appeared unaffected. In addition, circulating IGF-I is reduced by 30–50% in affected male but not in female mice. In addition to STAT5b, other transcriptions factors are related with body growth. This is exemplified by the glucocorticoid receptor (GR), which is a critical coactivator of STAT5b in liver [52], or by interactions between estradiol (E2)/estrogen receptor alpha (ERα) signaling and STAT5 [53]. In addition to endocrine actions, paracrine involvement of STAT5a/b in the effects of GH on muscle is also evident in the loss of muscle IGF-I transcripts and mass seen with muscle-specific deletion of STAT5a/b [54].

Advertisement

6. SOCS2 and body growth

The importance of SOCS2 in the negative regulation of body growth through inhibition of GHR-JAK2-STAT5b signaling was further demonstrated using genetically modified mice. Thus, the SOCS2-/- overgrowth phenotype is fully dependent on GH and can be rescued by inhibiting GH expression in these mice by crossing them with Ghrhrlit/lit mice that have no circulating GH. Both the double-knockout mice and the Ghrhrlit/lit mice exhibited a similar 60% growth retardation. Furthermore, administration of GH to these knockout mice caused an increase of growth to a size indistinguishable from SOCS2-/- mice [21, 55]. Similarly, mice resulting from crossing SOCS2-/- with STAT5b-/- mice show growth rates close to wild-type mice [56]. Similar phenotypes to the SOCS2-/- mice have been observed in high-growth (hg) mice, a phenotype that occurs following spontaneous mutation in mouse chromosome 10 [57]. However, in contrast to SOCS2-/-, hg mice have higher plasma IGF-I levels [57]. Surprisingly, overexpression of SOCS2 results in a similar phenotype to SOCS2-/- mice [34, 58], which suggests that the effects of SOCS2 on GH signaling are dose-dependent, with dual effects [16, 58]. It has been proposed that at physiological levels, SOCS2 inhibits GH signaling, by promoting GHR degradation, but at higher doses, it inhibits signaling of other, more potent GH inhibiting SOCS (i.e., SOCS1 and 3) [34, 58]. This could be through association with SOCS3 binding sites on the GHR, thus blocking SOCS3 action, or by binding the other SOCS themselves and suppressing them through proteasomal degradation [22]. The validity of these mechanisms has been questioned [59] and their physiological relevance remains uncertain. A more likely explanation of the effects created by SOCS2 overexpression would be a disruption of its E3 ligase activity by sequestering components of the multimeric E3 ligase away from the GHR.

Several studies have demonstrated that polymorphisms in the GH/IGF-I/SOCS2 system can also modulate the efficacy of GH treatment in humans. In humans with GH insensitivity due to a GHR defects, growth retardation, and reduced bone density, which are the result of IGF-I deficiency, are observed [60]. More recently, abnormalities of STAT5b, the IGF-IR gene itself, and the binding proteins that influence IGF-I bioavailability at the tissue level have all been reported to be associated with a variable extent of short stature in humans [12]. The effects of GH treatment on growth can also be influenced by polymorphisms on GHR or IGFBP3 genes and by their interactions among polymorphisms [6164]. Genetic polymorphisms in the SOCS2 gene have been associated with adult height variation in healthy individuals [13, 14, 65]. Recently, Braz et al. observed that SOCS2 polymorphism and its interaction with polymorphisms in GHR and IGFBP3 loci influenced the adult height of children with Turner syndrome and GHD (Growth Hormone Deficiency) after GH therapy [66]. Moreover, a SNP (Single Nucleotide Polymorphism) in the SOCS2 gene was reported associated to increased pubertal height in a Finnish cohort, supporting the role of SOCS2 in body growth in humans [67].

Advertisement

7. SOCS2 actions in bone and skeletal muscle

The overgrowth phenotype of the SOCS2 null (SOCS2-/-) mice [68] led to the confirmation that SOCS2 is a key effector of GH/IGF-I axis, in line with the anabolic role of GH on the skeleton [69, 70]. An interaction between SOCS2 and GH signaling in regulating body growth is consistent with the temporal increased expression of the GHR and the overgrowth of SOCS2-/- mice [68], with both occurring at around 3 weeks of age [68, 71]. Adult male SOCS2-/- mice are 40% heavier than their WT littermates while adult females reach the same size as the WT males [68]. Notably, the increased body weight of SOCS2-/- is not a result of any increase in fatty tissue but rather a proportional increase in size of most internal organs, muscle, and bone. SOCS2-/- mice have increased body length with longer longitudinal bones (femur, tibia, radius, and humerus) [68, 72]. No alterations to the growth plate were noted in the first description of SOCS2-/- mice [68]. Later investigations, however, found that epiphyseal chondrocytes express SOCS2 and growth plates from SOCS2-/- mice were enlarged with wider proliferative and hypertrophic zones. These findings were associated with an increased long bone length [72]. Recently, microtomography (μCT) analysis at 7 weeks of age showed that SOCS2-/- mice have increased bone mass (i.e., increased bone volume, trabecular number, and trabecular thickness), although these mice exhibit no difference in bone mineral density (BMD) compared to WT littermates [72]. In contrast, others authors have described lower trabecular and cortical bone mineral density in SOCS2-/- mice (at 4 and 15 weeks of age) as well as reduced cortical cross-sectional area and cortical thickness (at 4 weeks of age) [73]. Interestingly, these studies found elevated serum levels of osteocalcin (a marker of bone growth) [72, 73] and TRAP5 (a marker of osteoclast number) [72], which would indicate increased bone turnover in SOCS2-/- mice [68, 72]. Although circulating IGF-I levels are normal in SOCS2-/- mice [68, 72], they have elevated IGF-I mRNA in some tissues (heart, lung, and spleen but not liver, bone, fat, and muscle). Therefore, it is likely that the increased bone growth and observed structural differences within SOCS2-/- growth plates are a direct consequence of altered SOCS2-mediated GH signaling at the growth plate [33, 72]. Recent studies of Dobie and coworkers support this hypothesis. Using ex vivo metatarsal cultures, they showed that GH was able to induce linear growth only in the absence of SOCS2 [74] via a mechanism that is independent of IGF-I.

In addition to increased bone length, enhanced GHR signaling by GH treatment or SOCS2 deficiency causes skeletal muscle enlargement [68]. The molecular effects of GH and SOCS2 on skeletal muscles are uncertain. GH actions on skeletal muscles are a consequence of its systemic metabolic effects but also in part mediated by hormone-induced changes in gene expression within the muscle, as demonstrated by studies that show GH-induced changes in gene expression, including SOCS2, in human [75, 76] and murine [77] skeletal muscles. SOCS2 also modulates signaling pathways in the muscle independently from GH. Using C2C12 mesenchymal precursor cells, Ouyang et al. have shown that SOCS2 interferes with myotube formation and favors the differentiation into osteoblast in a process that, although not fully understood, would require the regulation of JunB [78]. In line with these observations, SOCS2 would also suppress myotube formation by inhibiting mitochondria biogenesis by interfering with the p38/ATF2 pathway [79]. Overall, these investigations highlight the importance of SOCS2 actions on bone and muscle development and growth through the modulation of multiple pathways, not restricted to GH signaling. Moreover, SOCS2 plays a key role as mediator of the interplay between sex steroids and GH signaling in these tissues.

Advertisement

8. Other functions of GH in the regulation of body weight

Disruption of GHR-STAT5-SOCS2 signaling pathway is also associated with metabolic disorders [17, 26, 51, 8084]. An inefficient GHR-JAK2-STAT5b signaling pathway results in fatty liver and adiposity in rodents and humans due to enhanced lipogenesis and reduced triglyceride secretion as well as reduced lipolysis [4, 80, 81]. This is supported by original findings showing that STAT5b null male mice become obese in later life [51] and that STAT5b deletion in a mature human was associated with obesity [85]. Therefore, GHR activated STAT5b plays a critical role in regulation of key enzymes involved in lipid and energy balance. Liver-specific GHR ablation leads to fatty liver because of reduced STAT5 activation despite normal plasma free fatty acid and minimal adiposity. Relevant to this review, agonists of liver X receptor (LXR), which cause hepatic steatosis [86], inhibit GH-STAT5b signaling [87]. This inhibition is mediated by SREBP1, a LXR target gene, through the downregulation of STAT5b gene transcription and stimulation of STAT5b protein degradation [87]. In contrast, ablation of SOCS2 in mice, which increased STAT5 signaling, protects them from high-fat diet-induced liver steatosis by increasing hepatic triglyceride secretion. As a result, these mice have increased peripheral fat accumulation both in adipose and muscle tissues. Although this is not associated with changes in systemic insulin sensitivity when mice are fed on a normal chow diet, under high-fat diet conditions, SOCS2-/- mice are glucose intolerant and insulin resistant and show increased expression of inflammatory cytokines [26]. The latter has been suggested to be a consequence of increased sensitivity of SOCS2-/- macrophages to lipopolysaccharide (LPS), leading to increased NFkB activation and inflammatory signaling in the liver despite reduced steatosis [26]. In contrast, SOCS2 deletion protected against streptozotocin-induced type I diabetes in adult male mice presumably by enhancing antiapoptotic actions of STAT5b [88]. Notably, SOCS2 can regulate inflammation by modulating the actions of other inflammatory cytokines [89]. The role of SOCS2 in the regulation of the inflammatory response seems to be independent of GH signaling. Furthermore, the effects of inflammatory cytokines on SOCS2 have been poorly investigated, with evidence that some interleukins induce SOCS2 gene expression only in specific cell types [89]. Thus, it has recently reported that in a sheep model, a point mutation in the SOCS2 gene not only resulted in higher body weight and size but also elevated leukocytes count in the milk as a sign of enhanced inflammatory response [90]. In addition, altered SOCS2 expression has also been associated with malignancies [84, 9193]. Therefore, how to target SOCS2-regulated pathways without causing negative side effects that systemic and chronic reduction in SOCS2 protein might cause is still a challenge.

Advertisement

9. SOCS2 mediates the cross-talk between steroids and GH

Until recently, most studies concerning the interaction among GH and steroids have been focused on the influence of sex steroids on gender-specific pituitary GH secretion that has a great impact on hepatic transcriptional regulation [17, 94]. However, a direct target of steroids may also occur because interaction with androgen receptor (AR), estrogen receptor alpha or GR, and the signaling pathways linked to these receptors are connected with lipid and glucose homeostasis [95] and tissue growth [96, 97]. The relationship between GH and sex steroids relative to body growth has been extensively studied, but it is not fully understood. Sex steroids can directly modulate pituitary GH secretion [94, 98] in a process that in men seems to require prior aromatization of testosterone into estrogen [99] but also indirectly through regulating liver IGF-1 production. Thus, hypogonadal children have reduced GH secretion [100, 101] and girls with precocious puberty show increased levels [102]. Additionally, sex steroids also exert GH-independent effects on growth [103, 104]. During puberty, children experience a growth spurt concomitant with increasing levels of gonadal steroids and GH secretions [98, 103]. This pubertal growth spurt has been attributed primarily to the actions of estrogens [105], acting directly on the growth plate cartilage inducing proliferation and finally promoting epiphyseal fusion [106]. Thus, in girls with Turner syndrome, hormone replacement therapy results in growth spurt [107]. Due to these effects on bone maturation, pharmacological doses of sex steroids have been used in prepubertal children with the nonpathological condition of constitutionally tall stature to limit their final height since 1950s [108, 109].

At the molecular level, increasing amount of evidences suggests that SOCS2 as a key mediator of the interplay between GH and steroid hormones. Both androgens and estrogens are able to induce the expression of SOCS2, which in turn limits GH signaling in cells from different tissue origins such as liver, breast, and prostate [84, 110]. Transcriptional induction of SOCS2 expression by sex steroids is mediated by the steroid receptors, AR, for androgens, and ERα, in the case of estrogens. This activation seems to be mediated by STAT5 [84] in a similar fashion to what happened with another steroid receptor, the GR, which acts as cofactor for STAT5-mediated transcription of SOCS2 after glucocorticoid stimulation [27]. Thus, direct upregulation of SOCS2 expression by steroid hormones would limit GH actions on target tissues. The liver, a major target tissue of GH, expresses both AR and ERα. Therefore, SOCS2 might play a central role in the modulation of GH signaling by androgens and estrogens, which defines the gender dimorphism in response to GH. In good agreement to the role of SOCS2 as mediator of sex steroids and GH signaling, SOCS2 KO mice show a less pronounced growth reduction after E2 treatment than their WT mates (unpublished observations). Recently, Bolamperti and colleagues described a novel SOCS2 regulation by estradiol in human osteoblasts [111]. Here, E2 induces GH signaling (STAT5 phosphorylation after treatment with GH) by inhibiting SOCS2 expression through a mechanism that involves proteasomal degradation of the protein but not genomic actions. The molecular characterization of this regulation, e.g., whether these effects are ER mediated, as well as the elucidation of this regulation as a general mechanism or whether it is restricted to osteoblast cells deserve further investigation. Overall, steroids in general and sex steroids in particular can modulate GH actions by controlling SOCS2 expression in several ways and in a tissue-specific manner. In the liver, SOCS2 induction by steroids would modulate central metabolism as well as influence IGF-I secretion that in turn affects GH secretion. In the bone, however, estrogens would potentiate GH actions by reducing the expression of SOCS2.

Advertisement

10. General conclusions

GH is the main regulator of somatic growth through pleiotropic actions on systemic metabolism and local actions on the bone growth plate. Relevant is the critical role of SOCS2 for the negative regulation of body growth. However, many aspects on the actions of SOCS2 in physiological and pathological models have yet to be understood. Particularly, more studies investigating the mechanisms by which SOCS2 regulates metabolism (e.g., lipid metabolism), insulin actions, malignancies, or GHR signaling at the growth plate are certainly needed. Notably, how to target the elements of the SOCS2-regulated pathways without causing negative side effects that systemic and chronic reduction in SOCS2 protein might cause is still a challenge. Finally, the consequences of long-term exposition to steroid compounds (particularly, sex hormones-related compounds) on normal development, as a consequence of their influence on GHR signaling, are largely unknown. Understanding this complex interaction in physiological and pathological states could contribute to prevent health damage and improve clinical management of patients with somatic growth disorders.

Acknowledgments

We thank all the authors that have made a contribution to the understanding of the negative regulation of GHR signaling. We apologize to those whose work deserves to be cited but unfortunately are not quoted because of space limitations. The research program in the author’s lab was supported by grants-in-aid from the Spanish Ministry of Economy and Competitiveness (MINECO) with the funding of European Regional Development Fund, European Social Fund, and Alfredo Martin-Reyes Foundation (Arehucas)—Canary Islands Foundation for Cancer Research (FICIC). D.I.-G. and A.F.-M. are supported by grants from the Novo Nordisk Foundation and the Danish Cancer Society.

References

  1. 1. Lichanska AM, Waters MJ: How growth hormone controls growth, obesity and sexual dimorphism. Trends Genet 2008, 24(1):41–47.
  2. 2. Moller N, Jorgensen JO: Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev 2009, 30(2):152–177.
  3. 3. Baik M, Yu JH, Hennighausen L: Growth hormone-STAT5 regulation of growth, hepatocellular carcinoma, and liver metabolism. Ann NY Acad Sci 2011, 1229:29–37.
  4. 4. 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):356–386.
  5. 5. Brooks AJ, Waters MJ: The growth hormone receptor: mechanism of activation and clinical implications. Nat Rev Endocrinol 2010, 6(9):515–525.
  6. 6. Carter-Su C, Schwartz J, Argetsinger LS: Growth hormone signaling pathways. Growth Horm IGF Res 2016, 28:11–15.
  7. 7. Vidal OM, Merino R, Rico-Bautista E, Fernandez-Perez L, Chia DJ, Woelfle J, Ono M, Lenhard B, Norstedt G, Rotwein P, et al.: In vivo transcript profiling and phylogenetic analysis identifies suppressor of cytokine signaling 2 as a direct signal transducer and activator of transcription 5b target in liver. Mol Endocrinol 2007, 21(1):293–311.
  8. 8. Cesena TI, Cui TX, Piwien-Pilipuk G, Kaplani J, Calinescu AA, Huo JS, Iniguez-Lluhi JA, Kwok R, Schwartz J: Multiple mechanisms of growth hormone-regulated gene transcription. Mol Genet Metab 2007, 90(2):126–133.
  9. 9. Wauthier V, Waxman DJ: Sex-specific early growth hormone response genes in rat liver. Mol Endocrinol 2008, 22(8):1962–1974.
  10. 10. Waxman DJ, O’Connor C: Growth hormone regulation of sex-dependent liver gene expression. Mol Endocrinol 2006, 20(11):2613–2629.
  11. 11. Rosenfeld RG, Kofoed E, Buckway C, Little B, Woods KA, Tsubaki J, Pratt KA, Bezrodnik L, Jasper H, Tepper A, et al.: Identification of the first patient with a confirmed mutation of the JAK-STAT system. Pediatr Nephrol 2005, 20(3):303–305.
  12. 12. Walenkamp MJ, Vidarsdottir S, Pereira AM, Karperien M, van Doorn J, van Duyvenvoorde HA, Breuning MH, Roelfsema F, Kruithof MF, van Dissel J, et al.: Growth hormone secretion and immunological function of a male patient with a homozygous STAT5b mutation. Eur J Endocrinol 2007, 156(2):155–165.
  13. 13. Weedon MN, Lango H, Lindgren CM, Wallace C, Evans DM, Mangino M, Freathy RM, Perry JR, Stevens S, Hall AS, et al.: Genome-wide association analysis identifies 20 loci that influence adult height. Nat Genet 2008, 40(5):575–583.
  14. 14. Gudbjartsson DF, Walters GB, Thorleifsson G, Stefansson H, Halldorsson BV, Zusmanovich P, Sulem P, Thorlacius S, Gylfason A, Steinberg S, et al.: Many sequence variants affecting diversity of adult human height. Nat Genet 2008, 40(5):609–615.
  15. 15. Rosenbloom AL: A half-century of studies of growth hormone insensitivity/Laron síndrome: a historical perspective. Growth Horm IGF Res 2016, 28:46–50.
  16. 16. Flores-Morales A, Greenhalgh CJ, Norstedt G, Rico-Bautista E: Negative regulation of growth hormone receptor signaling. Mol Endocrinol 2006, 20(2):241–253.
  17. 17. Mode A, Gustafsson JA: Sex and the liver—a journey through five decades. Drug Metab Rev 2006, 38(1–2):197–207.
  18. 18. da Silva Almeida AC, Strous GJ, van Rossum AG: BetaTrCP controls GH receptor degradation via two different motifs. Mol Endocrinol 2012, 26(1):165–177.
  19. 19. Rico-Bautista E, Flores-Morales A, Fernandez-Perez L: Suppressor of cytokine signaling (SOCS) 2, a protein with multiple functions. Cytokine Growth Factor Rev 2006, 17(6):431–439.
  20. 20. Linossi EM, Babon JJ, Hilton DJ, Nicholson SE: Suppression of cytokine signaling: the SOCS perspective. Cytokine Growth Factor Rev 2013, 24(3):241–248.
  21. 21. Greenhalgh CJ, Rico-Bautista E, Lorentzon M, Thaus AL, Morgan PO, Willson TA, Zervoudakis P, Metcalf D, Street I, Nicola NA, et al.: SOCS2 negatively regulates growth hormone action in vitro and in vivo. J Clin Invest 2005, 115(2):397–406.
  22. 22. Uyttendaele I, Lemmens I, Verhee A, De Smet AS, Vandekerckhove J, Lavens D, Peelman F, Tavernier J: Mammalian protein-protein interaction trap (MAPPIT) analysis of STAT5, CIS, and SOCS2 interactions with the growth hormone receptor. Mol Endocrinol 2007, 21(11):2821–2831.
  23. 23. Zhou W, Wei W, Sun Y: Genetically engineered mouse models for functional studies of SKP1-CUL1-F-box-protein (SCF) E3 ubiquitin ligases. Cell Res 2013, 23(5):599–619.
  24. 24. Vesterlund M, Zadjali F, Persson T, Nielsen ML, Kessler BM, Norstedt G, Flores-Morales A: The SOCS2 ubiquitin ligase complex regulates growth hormone receptor levels. PLoS One 2011, 6(9):e25358.
  25. 25. da Silva Almeida AC, Hocking HG, Boelens R, Strous GJ, van Rossum AG: BetaTrCP interacts with the ubiquitin-dependent endocytosis motif of the GH receptor in an unconventional manner. Biochem J 2013, 453(2):291–301.
  26. 26. Zadjali F, Santana-Farre R, Vesterlund M, Carow B, Mirecki-Garrido M, Hernandez-Hernandez I, Flodstrom-Tullberg M, Parini P, Rottenberg M, Norstedt G, et al.: SOCS2 deletion protects against hepatic steatosis but worsens insulin resistance in high-fat-diet-fed mice. FASEB J 2012, 26(8):3282–3291.
  27. 27. Tollet-Egnell P, Flores-Morales A, Stavreus-Evers A, Sahlin L, Norstedt G: Growth hormone regulation of SOCS-2, SOCS-3, and CIS messenger ribonucleic acid expression in the rat. Endocrinology 1999, 140(8):3693–3704.
  28. 28. Greenhalgh CJ, Alexander WS: Suppressors of cytokine signalling and regulation of growth hormone action. Growth Horm IGF Res 2004, 14(3):200–206.
  29. 29. Michaylira CZ, Simmons JG, Ramocki NM, Scull BP, McNaughton KK, Fuller CR, Lund PK: Suppressor of cytokine signaling-2 limits intestinal growth and enterotrophic actions of IGF-I in vivo. Am J Physiol Gastrointest Liver Physiol 2006, 291(3):G472–G481.
  30. 30. Adams TE, Hansen JA, Starr R, Nicola NA, Hilton DJ, Billestrup N: Growth hormone preferentially induces the rapid, transient expression of SOCS-3, a novel inhibitor of cytokine receptor signaling. J Biol Chem 1998, 273(3):1285–1287.
  31. 31. Hansen JA, Lindberg K, Hilton DJ, Nielsen JH, Billestrup N: Mechanism of inhibition of growth hormone receptor signaling by suppressor of cytokine signaling proteins. Mol Endocrinol 1999, 13(11):1832–1843.
  32. 32. Yasukawa H, Sasaki A, Yoshimura A: Negative regulation of cytokine signaling pathways. Annu Rev Immunol 2000, 18:143–164.
  33. 33. Alexander WS, Starr R, Metcalf D, Nicholson SE, Farley A, Elefanty AG, Brysha M, Kile BT, Richardson R, Baca M, et al.: Suppressors of cytokine signaling (SOCS): negative regulators of signal transduction. J Leukoc Biol 1999, 66(4):588–592.
  34. 34. Turnley AM: Role of SOCS2 in growth hormone actions. Trends Endocrinol Metab 2005, 16(2):53–58.
  35. 35. Leung KC, Waters MJ, Markus I, Baumbach WR, Ho KK: Insulin and insulin-like growth factor-I acutely inhibit surface translocation of growth hormone receptors in osteoblasts: a novel mechanism of growth hormone receptor regulation. Proc Natl Acad Sci USA 1997, 94(21):11381–11386.
  36. 36. De Rocca Serra-Nedelec A, Edouard T, Treguer K, Tajan M, Araki T, Dance M, Mus M, Montagner A, Tauber M, Salles JP, et al.: Noonan syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1 release via growth hormone-induced ERK hyperactivation, which contributes to short stature. Proc Natl Acad Sci USA 2012, 109(11):4257–4262.
  37. 37. Ehlting C, Bohmer O, Hahnel MJ, Thomas M, Zanger UM, Gaestel M, Knoefel WT, Schulte Am Esch J, Haussinger D, Bode JG: Oncostatin M regulates SOCS3 mRNA stability via the MEK-ERK1/2-pathway independent of p38(MAPK)/MK2. Cell Signal 2015, 27(3):555–567.
  38. 38. Ram PA, Waxman DJ: Interaction of growth hormone-activated STATs with SH2-containing phosphotyrosine phosphatase SHP-1 and nuclear JAK2 tyrosine kinase. J Biol Chem 1997, 272(28):17694–17702.
  39. 39. Hackett RH, Wang YD, Sweitzer S, Feldman G, Wood WI, Larner AC: Mapping of a cytoplasmic domain of the human growth hormone receptor that regulates rates of inactivation of Jak2 and Stat proteins. J Biol Chem 1997, 272(17):11128–11132.
  40. 40. Gu F, Dube N, Kim JW, Cheng A, Ibarra-Sanchez Mde J, Tremblay ML, Boisclair YR: Protein tyrosine phosphatase 1B attenuates growth hormone-mediated JAK2-STAT signaling. Mol Cell Biol 2003, 23(11):3753–3762.
  41. 41. Pasquali C, Curchod ML, Walchli S, Espanel X, Guerrier M, Arigoni F, Strous G, Hooft van Huijsduijnen R: Identification of protein tyrosine phosphatases with specificity for the ligand-activated growth hormone receptor. Mol Endocrinol 2003, 17(11):2228–2239.
  42. 42. Le Roith D, Bondy C, Yakar S, Liu JL, Butler A: The somatomedin hypothesis: 2001. Endocr Rev 2001, 22(1):53–74.
  43. 43. Kaplan SA, Cohen P: The somatomedin hypothesis 2007: 50 years later. J Clin Endocrinol Metab 2007, 92(12):4529–4535.
  44. 44. Lupu F, Terwilliger JD, Lee K, Segre GV, Efstratiadis A: Roles of growth hormone and insulin-like growth factor 1 in mouse postnatal growth. Dev Biol 2001, 229(1):141–162.
  45. 45. Giustina A, Mazziotti G, Canalis E: Growth hormone, insulin-like growth factors, and the skeleton. Endocr Rev 2008, 29(5):535–559.
  46. 46. Hutchison MR, Bassett MH, White PC: Insulin-like growth factor-I and fibroblast growth factor, but not growth hormone, affect growth plate chondrocyte proliferation. Endocrinology 2007, 148(7):3122–3130.
  47. 47. Yakar S, Liu JL, Stannard B, Butler A, Accili D, Sauer B, LeRoith D: Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci USA 1999, 96(13):7324–7329.
  48. 48. Liu JL, Yakar S, LeRoith D: Conditional knockout of mouse insulin-like growth factor-1 gene using the Cre/loxP system. Proc Soc Exp Biol Med 2000, 223(4):344–351.
  49. 49. Stratikopoulos E, Szabolcs M, Dragatsis I, Klinakis A, Efstratiadis A: The hormonal action of IGF1 in postnatal mouse growth. Proc Natl Acad Sci USA 2008, 105(49):19378–19383.
  50. 50. Le Roith D, Scavo L, Butler A: What is the role of circulating IGF-I? Trends Endocrinol Metab 2001, 12(2):48–52.
  51. 51. Udy GB, Towers RP, Snell RG, Wilkins RJ, Park SH, Ram PA, Waxman DJ, Davey HW: Requirement of STAT5b for sexual dimorphism of body growth rates and liver gene expression. Proc Natl Acad Sci USA 1997, 94(14):7239–7244.
  52. 52. Mueller KM, Themanns M, Friedbichler K, Kornfeld JW, Esterbauer H, Tuckermann JP, Moriggl R: Hepatic growth hormone and glucocorticoid receptor signaling in body growth, steatosis and metabolic liver cancer development. Mol Cell Endocrinol 2012, 361(1–2):1–11.
  53. 53. Bjornstrom L, Sjoberg M: Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 2005, 19(4):833–842.
  54. 54. Klover P, Hennighausen L: Postnatal body growth is dependent on the transcription factors signal transducers and activators of transcription 5a/b in muscle: a role for autocrine/paracrine insulin-like growth factor I. Endocrinology 2007, 148(4):1489–1497.
  55. 55. Leroith D, Nissley P: Knock your SOCS off! J Clin Invest 2005, 115(2):233–236.
  56. 56. Greenhalgh CJ, Bertolino P, Asa SL, Metcalf D, Corbin JE, Adams TE, Davey HW, Nicola NA, Hilton DJ, Alexander WS: Growth enhancement in suppressor of cytokine signaling 2 (SOCS-2)-deficient mice is dependent on signal transducer and activator of transcription 5b (STAT5b). Mol Endocrinol 2002, 16(6):1394–1406.
  57. 57. Horvat S, Medrano JF: Lack of Socs2 expression causes the high-growth phenotype in mice. Genomics 2001, 72(2):209–212.
  58. 58. Greenhalgh CJ, Metcalf D, Thaus AL, Corbin JE, Uren R, Morgan PO, Fabri LJ, Zhang JG, Martin HM, Willson TA, et al.: Biological evidence that SOCS-2 can act either as an enhancer or suppressor of growth hormone signaling. J Biol Chem 2002, 277(43):40181–40184.
  59. 59. Kiu H, Greenhalgh CJ, Thaus A, Hilton DJ, Nicola NA, Alexander WS, Roberts AW: Regulation of multiple cytokine signalling pathways by SOCS3 is independent of SOCS2. Growth Factors 2009, 27(6):384–393.
  60. 60. Parks JS, Brown MR, Faase ME: The spectrum of growth-hormone insensitivity. J Pediatr 1997, 131(1 Pt 2):S45–S50.
  61. 61. Wassenaar MJ, Dekkers OM, Pereira AM, Wit JM, Smit JW, Biermasz NR, Romijn JA: Impact of the exon 3-deleted growth hormone (GH) receptor polymorphism on baseline height and the growth response to recombinant human GH therapy in GH-deficient (GHD) and non-GHD children with short stature: a systematic review and meta-analysis. J Clin Endocrinol Metab 2009, 94(10):3721–3730.
  62. 62. Renehan AG, Solomon M, Zwahlen M, Morjaria R, Whatmore A, Audi L, Binder G, Blum W, Bougneres P, Santos CD, et al.: Growth hormone receptor polymorphism and growth hormone therapy response in children: a Bayesian meta-analysis. Am J Epidemiol 2012, 175(9):867–877.
  63. 63. Braz AF, Costalonga EF, Montenegro LR, Trarbach EB, Antonini SR, Malaquias AC, Ramos ES, Mendonca BB, Arnhold IJ, Jorge AA: The interactive effect of GHR-exon 3 and -202 A/C IGFBP3 polymorphisms on rhGH responsiveness and treatment outcomes in patients with Turner syndrome. J Clin Endocrinol Metab 2012, 97(4):E671–E677.
  64. 64. Costalonga EF, Antonini SR, Guerra G, Jr., Coletta RR, Franca MM, Braz AF, Mendonca BB, Arnhold IJ, Jorge AA: Growth hormone pharmacogenetics: the interactive effect of a microsatellite in the IGF1 promoter region with the GHR-exon 3 and -202 A/C IGFBP3 variants on treatment outcomes of children with severe GH deficiency. Pharmacogenomics J 2012, 12(5):439–445.
  65. 65. Chan Y, Holmen OL, Dauber A, Vatten L, Havulinna AS, Skorpen F, Kvaloy K, Silander K, Nguyen TT, Willer C, et al.: Common variants show predicted polygenic effects on height in the tails of the distribution, except in extremely short individuals. PLoS Genet 2011, 7(12):e1002439.
  66. 66. Braz AF, Costalonga EF, Trarbach EB, Scalco RC, Malaquias AC, Guerra-Junior G, Antonini SR, Mendonca BB, Arnhold IJ, Jorge AA: Genetic predictors of long-term response to growth hormone (GH) therapy in children with GH deficiency and Turner syndrome: the influence of a SOCS2 polymorphism. J Clin Endocrinol Metab 2014, 99(9):E1808–E1813.
  67. 67. Sovio U, Bennett AJ, Millwood IY, Molitor J, O’Reilly PF, Timpson NJ, Kaakinen M, Laitinen J, Haukka J, Pillas D, et al.: Genetic determinants of height growth assessed longitudinally from infancy to adulthood in the northern Finland birth cohort 1966. PLoS Genet 2009, 5(3):e1000409.
  68. 68. Metcalf D, Greenhalgh CJ, Viney E, Willson TA, Starr R, Nicola NA, Hilton DJ, Alexander WS: Gigantism in mice lacking suppressor of cytokine signalling-2. Nature 2000, 405(6790):1069–1073.
  69. 69. Ohlsson C, Bengtsson BA, Isaksson OG, Andreassen TT, Slootweg MC: Growth hormone and bone. Endocr Rev 1998, 19(1):55–79.
  70. 70. Andreassen TT, Oxlund H: The effects of growth hormone on cortical and cancellous bone. J Musculoskelet Neuronal Interact 2001, 2(1):49–58.
  71. 71. Metcalf D, Alexander WS, Elefanty AG, Nicola NA, Hilton DJ, Starr R, Mifsud S, Di Rago L: Aberrant hematopoiesis in mice with inactivation of the gene encoding SOCS-1. Leukemia 1999, 13(6):926–934.
  72. 72. Macrae VE, Horvat S, Pells SC, Dale H, Collinson RS, Pitsillides AA, Ahmed SF, Farquharson C: Increased bone mass, altered trabecular architecture and modified growth plate organization in the growing skeleton of SOCS2 deficient mice. J Cell Physiol 2009, 218(2):276–284.
  73. 73. Lorentzon M, Greenhalgh CJ, Mohan S, Alexander WS, Ohlsson C: Reduced bone mineral density in SOCS-2-deficient mice. Pediatr Res 2005, 57(2):223–226.
  74. 74. Dobie R, Ahmed SF, Staines KA, Pass C, Jasim S, MacRae VE, Farquharson C: Increased linear bone growth by GH in the absence of SOCS2 is independent of IGF-1. J Cell Physiol 2015, 230(11):2796–2806.
  75. 75. Vestergaard PF, Vendelbo MH, Pedersen SB, Juul A, Ringgard S, Moller N, Jessen N, Jorgensen JO: GH signaling in skeletal muscle and adipose tissue in healthy human subjects: impact of gender and age. Eur J Endocrinol 2014, 171(5):623–631.
  76. 76. Clasen BF, Poulsen MM, Escande C, Pedersen SB, Moller N, Chini EN, Jessen N, Jorgensen JO: Growth hormone signaling in muscle and adipose tissue of obese human subjects: associations with measures of body composition and interaction with resveratrol treatment. J Clin Endocrinol Metab 2014, 99(12):E2565–E2573.
  77. 77. Resmini E, Morte B, Sorianello E, Gallardo E, de Luna N, Illa I, Zorzano A, Bernal J, Webb SM: Identification of novel GH-regulated genes in C2C12 cells. Horm Metab Res 2011, 43(13):919–930.
  78. 78. Ouyang X, Fujimoto M, Nakagawa R, Serada S, Tanaka T, Nomura S, Kawase I, Kishimoto T, Naka T: SOCS-2 interferes with myotube formation and potentiates osteoblast differentiation through upregulation of JunB in C2C12 cells. J Cell Physiol 2006, 207(2):428–436.
  79. 79. Gan L, Liu Z, Zhang Z, Yang X, Liu J, Sun C: SOCS2 inhibited mitochondria biogenesis via inhibiting p38 MAPK/ATF2 pathway in C2C12 cells. Mol Biol Rep 2014, 41(2):627–637.
  80. 80. Barclay JL, Nelson CN, Ishikawa M, Murray LA, Kerr LM, McPhee TR, Powell EE, Waters MJ: GH-dependent STAT5 signaling plays an important role in hepatic lipid metabolism. Endocrinology 2011, 152(1):181–192.
  81. 81. Sos BC, Harris C, Nordstrom SM, Tran JL, Balazs M, Caplazi P, Febbraio M, Applegate MA, Wagner KU, Weiss EJ: Abrogation of growth hormone secretion rescues fatty liver in mice with hepatocyte-specific deletion of JAK2. J Clin Invest 2011, 121(4):1412–1423.
  82. 82. Fan Y, Fang X, Tajima A, Geng X, Ranganathan S, Dong H, Trucco M, Sperling MA: Evolution of hepatic steatosis to fibrosis and adenoma formation in liver-specific growth hormone receptor knockout mice. Front Endocrinol (Lausanne) 2014, 5:218.
  83. 83. Klover P, Chen W, Zhu BM, Hennighausen L: Skeletal muscle growth and fiber composition in mice are regulated through the transcription factors STAT5a/b: linking growth hormone to the androgen receptor. FASEB J 2009, 23(9):3140–3148.
  84. 84. Iglesias-Gato D, Chuan YC, Wikstrom P, Augsten S, Jiang N, Niu Y, Seipel A, Danneman D, Vermeij M, Fernandez-Perez L, et al.: SOCS2 mediates the cross talk between androgen and growth hormone signaling in prostate cancer. Carcinogenesis 2014, 35(1):24–33.
  85. 85. Vidarsdottir S, Walenkamp MJ, Pereira AM, Karperien M, van Doorn J, van Duyvenvoorde HA, White S, Breuning MH, Roelfsema F, Kruithof MF, et al.: Clinical and biochemical characteristics of a male patient with a novel homozygous STAT5b mutation. J Clin Endocrinol Metab 2006, 91(9):3482–3485.
  86. 86. Hong C, Tontonoz P: Liver X receptors in lipid metabolism: opportunities for drug discovery. Nat Rev Drug Discov 2014, 13(6):433–444.
  87. 87. Zadjali F, Santana-Farre R, Mirecki-Garrido M, Ellis E, Norstedt G, Fernandez-Perez L, Flores-Morales A: Liver X receptor agonist downregulates growth hormone signaling in the liver. Horm Mol Biol Clin Investig 2011, 8(2):471–478.
  88. 88. Al Kharusi A, Mirecki-Garrido M, Ma Z, Zadjali F, Flores-Morales A, Nystrom T, Castrillo A, Bjorklund A, Norstedt G, Fernandez-Perez L. Suppressor of cytokine signaling 2 (SOCS2) deletion protects against multiple low dose-streptozotocin-induced type I diabetes in adult male mice. Horm Mol Biol Clin Investig 2016, 26(1):67–76.
  89. 89. Galic S, Sachithanandan N, Kay TW, Steinberg GR: Suppressor of cytokine signalling (SOCS) proteins as guardians of inflammatory responses critical for regulating insulin sensitivity. Biochem J 2014, 461(2):177–188.
  90. 90. Rupp R, Senin P, Sarry J, Allain C, Tasca C, Ligat L, Portes D, Woloszyn F, Bouchez O, Tabouret G, et al.: Apoint mutation in suppressor of cytokine signalling 2 (Socs2) increases the susceptibility to inflammation of the mammary gland while associated with higher body weight and size and higher milk production in a sheep model. PLoS Genet 2015, 11(12):e1005629.
  91. 91. Vitali C, Bassani C, Chiodoni C, Fellini E, Guarnotta C, Miotti S, Sangaletti S, Fuligni F, De Cecco L, Piccaluga PP, et al.: SOCS2 controls proliferation and stemness of hematopoietic cells under stress conditions and its deregulation marks unfavorable acute leukemias. Cancer Res 2015, 75(11):2387–2399.
  92. 92. Newton VA, Ramocki NM, Scull BP, Simmons JG, McNaughton K, Lund PK: Suppressor of cytokine signaling-2 gene disruption promotes Apc(Min/+) tumorigenesis and activator protein-1 activation. Am J Pathol 2010, 176(5):2320–2332.
  93. 93. Michaylira CZ, Ramocki NM, Simmons JG, Tanner CK, McNaughton KK, Woosley JT, Greenhalgh CJ, Lund PK: Haplotype insufficiency for suppressor of cytokine signaling-2 enhances intestinal growth and promotes polyp formation in growth hormone-transgenic mice. Endocrinology 2006, 147(4):1632–1641.
  94. 94. Kerrigan JR, Rogol AD: The impact of gonadal steroid hormone action on growth hormone secretion during childhood and adolescence. Endocr Rev 1992, 13(2):281–298.
  95. 95. Mauvais-Jarvis F: Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity. Trends Endocrinol Metab 2011, 22(1):24–33.
  96. 96. Fisher B, Gunduz N, Saffer EA, Zheng S: Relation of estrogen and its receptor to rat liver growth and regeneration. Cancer Res 1984, 44(6):2410–2415.
  97. 97. Vidal O, Lindberg MK, Hollberg K, Baylink DJ, Andersson G, Lubahn DB, Mohan S, Gustafsson JA, Ohlsson C: Estrogen receptor specificity in the regulation of skeletal growth and maturation in male mice. Proc Natl Acad Sci USA 2000, 97(10):5474–5479.
  98. 98. Meinhardt UJ, Ho KK: Modulation of growth hormone action by sex steroids. Clin Endocrinol (Oxf) 2006, 65(4):413–422.
  99. 99. Weissberger AJ, Ho KK: Activation of the somatotropic axis by testosterone in adult males: evidence for the role of aromatization. J Clin Endocrinol Metab 1993, 76(6):1407–1412.
  100. 100. Giustina A, Scalvini T, Tassi C, Desenzani P, Poiesi C, Wehrenberg WB, Rogol AD, Veldhuis JD: Maturation of the regulation of growth hormone secretion in young males with hypogonadotropic hypogonadism pharmacologically exposed to progressive increments in serum testosterone. J Clin Endocrinol Metab 1997, 82(4):1210–1219.
  101. 101. Jospe N, Orlowski CC, Furlanetto RW: Comparison of transdermal and oral estrogen therapy in girls with Turner’s syndrome. J Pediatr Endocrinol Metab 1995, 8(2):111–116.
  102. 102. Mansfield MJ, Rudlin CR, Crigler JF, Jr., Karol KA, Crawford JD, Boepple PA, Crowley WF, Jr.: Changes in growth and serum growth hormone and plasma somatomedin-C levels during suppression of gonadal sex steroid secretion in girls with central precocious puberty. J Clin Endocrinol Metab 1988, 66(1):3–9.
  103. 103. Perry RJ, Farquharson C, Ahmed SF: The role of sex steroids in controlling pubertal growth. Clin Endocrinol (Oxf) 2008, 68(1):4–15.
  104. 104. Venken K, Schuit F, Van Lommel L, Tsukamoto K, Kopchick JJ, Coschigano K, Ohlsson C, Moverare S, Boonen S, Bouillon R, et al.: Growth without growth hormone receptor: estradiol is a major growth hormone-independent regulator of hepatic IGF-I synthesis. J Bone Miner Res 2005, 20(12):2138–2149.
  105. 105. Drop SL, De Waal WJ, De Muinck Keizer-Schrama SM: Sex steroid treatment of constitutionally tall stature. Endocr Rev 1998, 19(5):540–558.
  106. 106. Weise M, De-Levi S, Barnes KM, Gafni RI, Abad V, Baron J: Effects of estrogen on growth plate senescence and epiphyseal fusion. Proc Natl Acad Sci USA 2001, 98(12):6871–6876.
  107. 107. Ross JL, Cassorla FG, Skerda MC, Valk IM, Loriaux DL, Cutler GB, Jr.: A preliminary study of the effect of estrogen dose on growth in Turner’s syndrome. N Engl J Med 1983, 309(18):1104–1106.
  108. 108. Goldzieher MA: Treatment of excessive growth in the adolescent female. J Clin Endocrinol Metab 1956, 16(2):249–252.
  109. 109. Frank GR: The role of estrogen in pubertal skeletal physiology: epiphyseal maturation and mineralization of the skeleton. Acta Paediatr 1995, 84(6):627–630.
  110. 110. Leung KC, Doyle N, Ballesteros M, Sjogren K, Watts CK, Low TH, Leong GM, Ross RJ, Ho KK: Estrogen inhibits GH signaling by suppressing GH-induced JAK2 phosphorylation, an effect mediated by SOCS-2. Proc Natl Acad Sci USA 2003, 100(3):1016–1021.
  111. 111. Bolamperti S, Mrak E, Moro G, Sirtori P, Fraschini G, Guidobono F, Rubinacci A, Villa I: 17beta-Estradiol positively modulates growth hormone signaling through the reduction of SOCS2 negative feedback in human osteoblasts. Bone 2013, 55(1):84–92.

Written By

Leandro Fernández-Pérez, Amilcar Flores-Morales, Borja Guerra, Juan C. Díaz-Chico and Diego Iglesias-Gato

Submitted: 10 November 2015 Reviewed: 14 June 2016 Published: 12 October 2016