Skip to main content
Reproductive Medicine and Biology logoLink to Reproductive Medicine and Biology
. 2024 Apr 3;23(1):e12575. doi: 10.1002/rmb2.12575

Molecular crosstalk between insulin‐like growth factors and follicle‐stimulating hormone in the regulation of granulosa cell function

Emily Hayes 1, Nicola Winston 2, Carlos Stocco 1,2,
PMCID: PMC10988955  PMID: 38571513

Abstract

Background

The last phase of folliculogenesis is driven by follicle‐stimulating hormone (FSH) and locally produced insulin‐like growth factors (IGFs), both essential for forming preovulatory follicles.

Methods

This review discusses the molecular crosstalk of the FSH and IGF signaling pathways in regulating follicular granulosa cells (GCs) during the antral‐to‐preovulatory phase.

Main findings

IGFs were considered co‐gonadotropins since they amplify FSH actions in GCs. However, this view is not compatible with data showing that FSH requires IGFs to stimulate GCs, that FSH renders GCs sensitive to IGFs, and that FSH signaling interacts with factors downstream of AKT to stimulate GCs. New evidence suggests that FSH and IGF signaling pathways intersect at several levels to regulate gene expression and GC function.

Conclusion

FSH and locally produced IGFs form a positive feedback loop essential for preovulatory follicle formation in all species. Understanding the mechanisms by which FSH and IGFs interact to control GC function will help design new interventions to optimize follicle maturation, perfect treatment of ovulatory defects, improve in vitro fertilization, and develop new contraceptive approaches.

Keywords: FSH, granulosa cells, insulin‐like growth factors, ovary, steroidogenesis


Crosstalk between insulin‐like growth factors and follicle‐stimulating hormone in the regulation of ovarian antral follicles.

graphic file with name RMB2-23-e12575-g006.jpg

1. INTRODUCTION

Over the last 4 decades, groundbreaking research has systematically enhanced our understanding of the intricate molecular interplay between insulin‐like growth factors (IGFs) and gonadotropin signaling involved in the regulation of ovarian function and female fertility. Meticulous in vitro studies involving granulosa cells (GCs) from humans, rodents, nonhuman primates, and farm animals, coupled with innovative genetic approaches, have unveiled IGFs as pivotal intraovarian regulators. IGFs wield unparalleled influence over critical facets of follicle dynamics, including growth, selection, atresia, cellular differentiation, steroidogenesis, oocyte maturation, and cumulus expansion. Furthermore, the discerned actions of IGFs have illuminated a profound interdependency with gonadotropins, underscoring their status as indispensable paracrine regulatory factors beyond their initially described role as “co‐gonadotropins.” The implications of these findings extend far beyond the realm of basic research, holding the promise of transformative insights into fertility treatments and reproductive health interventions. Recent outstanding reviews have covered the roles of IGFs in ovarian development and their effects on reproductive tissues in general. 1 , 2 This review provides a comprehensive account of the effects of IGFs and the molecular interaction of IGF and follicle‐stimulating hormone (FSH) signaling in GCs.

2. FOLLICULOGENESIS OVERVIEW

The ovary fosters the development and maturation of the female gamete, the oocyte. The ovarian functional unit is the follicle, which consists of an oocyte surrounded by one or several layers of GCs enclosed by a basal lamina. The development of the ovarian follicle to the point at which a mature oocyte(s) is ovulated is termed folliculogenesis, a process that in humans lasts up to 1 year. 3 It starts with primordial follicle activation, which involves the transformation of squamous GCs into cuboidal cells to form primary follicles. In primary follicles, GCs start to proliferate slowly, forming several layers of cells to form secondary follicles. Secondary follicles acquire a layer of highly steroidogenic cells around the basal lamina known as the theca layer. Blood vessels develop within the theca layer, allowing systemic hormones to regulate follicle growth.

Further follicle development is driven by FSH, which is essential for the formation of antral follicles and, ultimately, their maturation to the preovulatory stage (Figure 1). During the last stages of maturation, GCs differentiate into mural cells, which produce high levels of estradiol, progesterone, and inhibins. These hormones coordinate oocyte maturation and ovulation with uterine receptivity. As we will describe in this review, the growth beyond the early antral stage and hormone production of follicles depends on the actions of FSH and IGFs. This review will focus on the molecular crosstalk of the FSH and IGF signaling pathways in the regulation of follicle maturation to the preovulatory stage.

FIGURE 1.

FIGURE 1

Follicle development. The oocyte is initially surrounded by a flattened layer of GCs in the primordial follicle. As the follicle progresses to the primary stage, the GCs change morphology to become a single layer of cuboidal cells. These GCs proliferate to form multiple layers in the secondary/preantral follicle. A fluid‐filled cavity then begins to form within the follicle, known as the antrum. In mono‐ovulatory species, a single follicle becomes dominant and progresses to the preovulatory stage. As the antrum grows, GCs differentiate into the mural GCs lining the wall of the follicle and cumulus GCs immediately surrounding the oocyte.

2.1. Overview of FSH signaling

FSH is produced by the gonadotrophs of the anterior pituitary and travels in blood to the gonads, where it acts exclusively on the GCs via binding to its receptor, the FSH receptor (FSHR). FSHR expression, and therefore FSH actions, are limited to GCs. 4 The FSHR is a G protein‐coupled receptor linked to the activation of stimulatory Gα. Gα activation stimulates adenylyl cyclase that catalyzes the conversion of ATP to cAMP. Thus, FSH activation of GCs increases intracellular cAMP, which in turn activates protein kinase A (PKA). One well‐known target of PKA is the cAMP response element‐binding protein (CREB), a transcription factor. CREB binds to cAMP response element sequences, boosting transcription of FSH‐regulated genes. 5 , 6 In addition to CREB, several transcription factors, including LRH1 and GATA4, participate in the regulation of GC steroidogenesis. 7 , 8 By inhibiting dual‐specific phosphatases, PKA mediates the activation of ERK, 9 a regulatory component of GC survival. PKA also promotes p38 MAPK and JNK signaling. 10 A well‐known intracellular target of the FSHR is AKT (acute transforming retrovirus thymoma protein kinase). 11 Downstream of AKT signaling are FOXO and HIF1α factors that are involved in GC proliferation. 12 , 13 , 14 This review will focus on the mechanisms involved in the regulation of AKT by FSH and IGFs (Figure 2). For a complete description of all signaling pathways and intracellular effectors of FSH in the GCs, see the following references. 15 , 16 , 17

FIGURE 2.

FIGURE 2

Simplified scheme of major FSH intracellular signaling. FSH binds and activates the FSHR, which stimulates adenylyl cyclase (AC) activity and increases intracellular cAMP. cAMP activates PKA, ERK, and AKT, which regulate the expression of FSH‐targeted genes.

2.2. Ovarian steroidogenesis

From the early antral to the preovulatory stage, the mural GCs and adjacent theca cells produce increasing levels of progestins, androgens, and estrogens. Mural GCs and theca cells cooperate to produce estradiol, which is critical for GC proliferation, follicle development, and the regulation of gonadotropin secretion. 18 Theca and GCs express the enzymes necessary to convert cholesterol into progesterone, 19 including steroid acute regulatory protein (StAR), which transports cholesterol into the mitochondria where cholesterol side‐chain cleavage enzyme (CYP11A1) catalyzes the first step in the steroidogenic pathway. Subsequent reactions by 3β‐Hydroxysteroid dehydrogenase/Δ5‐4 isomerase (3βHSD) leads to the production of progesterone. GCs are unable to convert progestins into androgens. However, theca cells specifically express cytochrome P450 Family 17 Subfamily A Member 1 (CYP17A1), which converts progesterone into androgen. Androgens then diffuse to the granulosa layer where aromatase (CYP19A1), which is exclusively expressed in GCs, converts androgens into estradiol 20 (Figure 3).

FIGURE 3.

FIGURE 3

Granulosa and theca cell steroidogenesis. A, androgens; BM, basal membrane; Ch, cholesterol; E, estrogens; P, progesterone.

3. INSULIN‐LIKE GROWTH FACTORS

IGF1 and IGF2 are polypeptides with structural similarity to insulin comprised of α and β chains linked by disulfide bonds. IGF1, initially known as somatomedin C, is produced mainly in the liver under the control of growth hormone (GH) and secreted into the systemic circulation. 21 However, IGF1 is also expressed locally in most tissues, including the ovary, exerting paracrine and autocrine actions. 22 IGF2 is considered the major growth factor during fetal development, but it is also expressed in various adult tissues, including the liver, although its actions in adult target tissues are largely unknown. 23

3.1. IGF receptors and signaling

The type 1 IGF receptor (IGF1R) mediates the stimulatory effects of IGF1 and IGF2. This receptor belongs to the family of receptor tyrosine kinases, and its activation triggers autophosphorylation of specific tyrosine residues in the receptor β‐subunits, thus promoting their tyrosine kinase activity (Figure 4). Tyrosine phosphorylation facilitates the binding of adaptors containing phosphotyrosine‐binding domains, such as insulin receptor substrate (IRS)1 and IRS2, which in turn are tyrosine phosphorylated (pY) by the receptor. 24

FIGURE 4.

FIGURE 4

IGFs signal through the IGF1R in GCs. IGF1 and IGF2 are both ligands for the IGF1R. After activation by either ligand, tyrosine residues in the intracellular region of the IGF1R are phosphorylated. This leads to the recruitment of insulin receptor substrate (IRS) that acts as a scaffold for proteins necessary for activating MAPK and PI3K/AKT pathways.

Class 1A phosphoinositide 3‐kinases (PI3Ks) are recruited to the membrane via the interaction of their p85 regulator subunit with pY‐IRSs 25 bringing the PI3K p110 catalytic subunit to the membrane where it converts phosphatidylinositol‐4,5‐bisphosphate (PIP2) to phosphatidylinositol‐3,4,5‐triphosphate (PIP3). PIP3 serves as a docking site for signaling proteins that include phosphoinositide‐dependent kinase 1 (PDK1). Phosphatase and tensin homolog 10 (PTEN) is a negative regulator of PI3K that converts PIP3 back to PIP2. AKT also binds to PIP3 causing the translocation of AKT to the plasma membrane and its colocalization with PDK1. Therefore, PIP3 brings PDK1 and AKT together allowing PDK1 to phosphorylate AKT at T308. 26 AKT becomes fully active after phosphorylation on S473 by the mammalian target of rapamycin complex 2 (mTORC2). 27 Although IGF1R activation has been shown to stimulate mitogen‐activated protein kinases (MAPK), 28 IGFs do not seem to be a significant stimulus for MAPK activation in rat and human GCs. 29 , 30 , 31

The type 2 IGF receptor (IGF2R) is a single‐chain polypeptide with large extracellular domains with high affinity for IGF2. 32 It is largely accepted that the IGF2R does not propagate intracellular signaling within cells. The primary role of the IGF2R is to mediate the internalization of IGF2 upon binding to the receptor. Additionally, the IGF2R can be released from the cell membrane, 33 which suggests it could block IGF2 actions in the intercellular space.

In addition to the expression of IGFs and their receptors, the bioavailability of IGFs dramatically changes during follicle growth. The bioavailability of IGFs is regulated by IGF‐binding proteins (IGFBPs) that bind IGF1 and IGF2 with high affinity and can both inhibit (mostly) and potentiate their actions. Since the primary focus of this review is the intracellular molecular crosstalk between FSH and IGFs, the regulation by IGFBPs will not be described. Recent excellent reviews on the role of IGFBPs in the ovary can be found in the references cited here. 34 , 35

3.2. Role of IGFs in GC function across species

3.2.1. Rats

Expression

The mRNA of IGF1 but not IGF2 was first detected in rat ovaries by Northern blot in 1987. 36 Later, in situ hybridization studies identified the GCs of healthy developing follicles as the primary site of IGF1 synthesis. 37 In large preovulatory follicles, IGF1 mRNA is found primarily on GCs lining the antrum and the cumulus cells but not in the mural GCs. 38 , 39 Thus, there is a progressive decrease in IGF1 expression in the GCs from the early antral to the preovulatory stage that starts in the mural GCs. The factors involved in the regulation of IGF1 expression are not known but seem to be gonadotropin‐independent since IGF1 mRNA levels remain unchanged after hypophysectomy with or without gonadotropin replacement. 38 Estradiol stimulates IGF1 in GCs but not in hepatocytes, indicating tissue‐specific regulation. 40 In vitro, IGF1 (but not IGF1R) mRNA expression rapidly decreases once GCs are placed in culture; however, its expression is maintained in the presence of insulin and dexamethasone. 41 In the corpus luteum, IGF1R is expressed at higher concentrations in early pregnancy, while IGF1 increases close to parturition 42 due to the stimulatory effects of prolactin. 42 , 43 Thus, estradiol, insulin, glucocorticoids, and prolactin may regulate IGF1 expression in rat GCs.

Earlier studies showed that GCs bind IGFs 44 and that binding is enhanced by FSH. 45 , 46 Accordingly, hypophysectomy decreases, whereas FSH treatment increases IGF1R expression without changes in IGF1 levels. 38 , 47 IGF1 and IGF1R expression is insensitive to growth hormone (GH) control 38 ; therefore, the ovarian IGF1 system is autonomous from circulating GH.

Paracrine roles of GC‐derived IGF1

IGF1 is restricted to the GC compartment as no IGF1 expression has been detected in rat theca cells. 37 , 40 However, early studies also showed that IGF1 can enhance basal and LH‐supported androgen biosynthesis in rat theca‐interstitial cells. 48 , 49 Similarly, in vivo, IGF1 acts synergistically with LH to induce hyperandrogenism in rats. 50 IGF1 stimulates CYP11A1 and 3βHSD in theca cells. 51 , 52 , 53 In contrast, IGF1 stimulates the expression of CYP17A1 only in the presence of LH. 54 Thus, IGF1 acts on the steroidogenesis of theca cells by augmenting and mimicking LH actions. IGF1 is also a potent stimulator of theca cell proliferation, increasing the number and proportion of steroidogenically active theca cells. 55 However, although IGF1R is highly abundant in the granulosa and luteal cells, it is not detected in rat theca cells. 38 , 56 Therefore, how IGF1 regulates theca cell function in rats remains unknown. Despite this, the evidence raises the possibility that IGF1 from GCs acts in a paracrine manner in the adjacent theca cells, enhancing ovarian androgen production to guarantee the estrogen requirements of the developing follicle (Figure 5).

FIGURE 5.

FIGURE 5

Intrafollicular autocrine and paracrine interactions between the oocyte, GCs, and theca cells mediated by the IGF system (see text for details).

Interestingly, the IGF1R is present in oocytes. 38 Therefore, mutual actions between the oocyte and GCs can be speculated in which the oocyte itself induces IGF1 expression in GCs, whereas the IGF1 produced by GCs could support the metabolic activity of the oocyte. Similar relationships appear to be at play in humans; see below. On the other hand, the knockdown of the IGF1R in the oocyte of mice has no consequences on fertility 57 ; therefore, the role of the IGF1R in oocyte physiology is unclear. Effects: Earlier in vitro reports demonstrated that low concentrations of IGF1 (~4.0 ng/mL) strongly synergize with FSH on the stimulation of progesterone, 58 , 59 , 60 estradiol, 60 , 61 and GC proliferation. 39 , 62 IGF1 also synergizes with FSH to stimulate luteinizing hormone (LH) receptor, 63 CYP19A1, 64 and cAMP production 60 , 65 as well as proteoglycan biosynthesis. 66 Eventually, it was shown that IGF1 enhances the FSH‐induced up‐regulation of StAR, CYP11A1, 3βHSD, and CYP19A1 expression. 31 , 67 However, IGF1 alone does not impact any of these genes. As an exception, IGF1 alone and synergistic with FSH stimulates inhibin 68 , 69 and cartilage link protein expression. 70 Interestingly, IGF1 increases LH receptor and FSHR mRNA stability but not gene transcription. 71 , 72 These experiments demonstrated that IGF1 has an important role in the process of GC differentiation from the preantral to the large antral follicle stage by potentiating FSH actions.

3.2.2. Mice

Expression

In the ovary of mice, the patterns of IGF1 and IGF1R expression are similar to those described in the rat. 73 Interestingly, IGF1 mRNA is present exclusively in a subset of follicles, while all follicles express IGF1R. 73 Later, it was shown that IGF1 is detected only in FSHR‐positive follicles. However, the specific pattern of IGF1 expression remains unaltered in the ovaries of FSH knockout (KO) mice. 74 Similarly, hypophysectomy in the presence or absence of FSH replacement does not affect IGF1 expression, 74 indicating that FSH also does not regulate IGF1 expression in mice.

Knockout mice

Most newborn IGF1 KO pups die after birth, though their survival rate is dependent on genetic background; about 10% of pups with the C57BL/6J background survive to adulthood, whereas 68% with the MF1 background survive. 75 Surviving IGF1 KO mice are infertile and exhibit delayed bone development and growth retardation, reaching only 30% of the typical adult weight. 76 In contrast, mice lacking IGF2 are fertile with normal ovarian function. 77

IGF1 KO female mice have hypoplastic gonads containing primordial, primary, and secondary antral follicles but only rare small antral follicles. 73 Moreover, IGF1 KO females fail to ovulate even after administration of gonadotropins, suggesting that ovarian dysfunction is the primary cause of infertility. 73 It was later shown that GCs of IGF1 KO mice have reduced glucose transporter 1 expression 78 and show impaired proliferation in the absence of apoptosis, although BAX expression is higher in these cells. 79 Thus, IGF1 KO mice demonstrated that IGF1 is required for antral follicle formation. Whether or not this effect of IGF1 is ovary‐specific was evaluated using conditional knockdown mice.

The IGF1R gene is essential for normal embryonic development and pup survival after birth. 75 Notably, the neonatal lethality of IGF1R KO mice is invariable and independent of the genetic background. However, female mice heterozygous for IGF1R (IGF1R+/−) and mice with only a reduction in IGF1R expression are fertile. 80 , 81 Similarly, most IGF2R KO mice die perinatally and exhibit overgrowth due to high levels of IGF2 in the circulation. The few IGF2R KO females that survive are infertile due to an imperforate vagina. 82 Strikingly, female mice lacking IGF1R and IGF2R are fertile but have small litter sizes. 82 The excess of IGF2 in the absence of IGF2R and IGF1R may partially rescue ovarian function by activating the insulin receptor (INSR) or another unidentified receptor, as proposed by Ludwig et al. 82

To explore the ovarian roles of IGFs without causing severe systemic defects, Baumgarten et al. 56 produced mice that lack IGF1R, specifically in the GCs (IGF1RGCKO). IGF1RGCKO female mice are viable and grow normally but are infertile; their ovaries lack antral follicles and do not ovulate even after treatment with exogenous gonadotropins. 56 GCs from IGF1RGCKO mice are prone to enter apoptosis and have a significant reduction in AKT activation and steroidogenic gene expression but normal levels of FSHR. 56 Ablation of INSR and/or IGF1R in the oocyte has no impact on fertility. 57 However, the knockdown of the INSR and IGF1R in GCs of preovulatory follicles leads to infertility due to a lack of luteal support. 83 Thus, only GCs require the IGF1R to undergo cell differentiation upon FSH stimulation in vivo.

What is the role of GH and liver‐derived IGF1 in ovarian function?

GH receptor (GHR) KO mice that lack endocrine IGF1 are subfertile, 84 , 85 and their ovaries contain atretic follicles and few preovulatory follicles. 84 IGF1 treatment of GHR KO mice increases the number of healthy antral follicles and decreases the percentage of atretic follicles but does not rescue fertility or the responsiveness to gonadotropins. 84 , 86 Similarly, IGF1 KO mice expressing IGF1 only in the liver can conceive and produce pups, although with difficulty, suggesting subfertility. 87 These findings suggest that GH regulates follicular growth independently of IGF1 and that in the absence of IGF1 production in the ovary, circulating (liver or exogenous) IGF1 can only partially sustain ovarian function.

3.2.3. Humans/primates

Expression

In humans, IGF1R is expressed in GCs of antral follicles and colocalizes with phosphorylated AKT and the FSHR. 88 IGF2 is highly expressed in these cells, but IGF1 is not detectable. 30 , 89 , 90 , 91 , 92 Consequently, the follicular fluid of dominant follicles contains 3‐ to 10‐fold more IGF2 than IGF1. 93 , 94 , 95 , 96 There is also a positive association between intrafollicular IGF2, follicular function, 94 , 96 and early embryo development. 97 As GH administration to pre‐menopausal women does not affect ovarian IGF2 expression, 98 and serum levels of IGF1 and IGF2 do not vary across the menstrual cycle, it is unlikely that systemic IGFs play any role in the regulation of human GCs. 96 High IGF2 production by human GCs explains the normal follicular development and fertility of Laron dwarfism patients with extremely low blood levels of IGF1. 99 , 100 Similarly, the high levels of IGF2 in primate follicles explain why exogenous administration of IGF1 does not affect follicle maturation in monkeys. 101 The preferential expression of IGF2 by dominant follicles suggests that IGF2 plays a critical role in follicle selection and maturation.

Effects

Although IGF1 is not expressed in human GCs, IGF2 and IGF1 stimulate steroid production by upregulating StAR, CYP11A1, and CYP19A1 expression in luteinized human GCs. 30 , 102 , 103 , 104 , 105 , 106 , 107 , 108 , 109 , 110 , 111 In preantral undifferentiated GCs, IGF2 also stimulates sex steroid synthesis, 112 , 113 , 114 increases survival, 115 and suppresses IGFBP1. 116 IGFs also act synergistically with FSH to stimulate steroid synthesis and proliferation in GCs from normal 30 , 107 and PCOS 117 follicles. Therefore, unlike rodents, IGF1 or IGF2 can stimulate aromatase expression in the absence of FSH in human GCs. 30

In human GCs, FSH and cAMP stimulate IGF2 expression 30 , 112 , 118 , 119 by activating promoter 3 of the IGF2 gene, 30 although the factors involved remain to be determined. Oocyte‐secreted factors (OSFs), such as growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), potentiate the stimulatory effect of FSH on IGF2 but have no effect by themselves. 120 , 121 These findings suggest an intricate interplay between OSFs, IGF2, and FSH is established in human follicles during GC differentiation. This interplay involves FSH stimulation of IGF2 that is strongly enhanced by OSFs. In turn, IGF2 autocrine actions activate the IGF1R in the GCs, leading to the activation of AKT, which is essential to promote the expression of IGF2 and CYP19A1 and sustaining proliferation. Thus, it is possible to postulate the formation of a positive loop interaction within the follicle, which is critical for follicle growth and possible selection of the dominant follicle (Figure 5).

Paracrine action of GC‐produced IGF2 in humans: In human primordial follicles, the IGF1R is restricted to the oocyte; however, in preantral follicles, the IGF1R is present in GCs and theca cells. 122 As follicles grow, IGF1R in granulosa and theca cells increases. 122 In small antral follicles, IGF1 and IGF2 mRNAs can also be found in theca cells. Although IGF1R is detectable only in GCs, IGF2R is expressed in granulosa and thecal cells. 90 In dominant follicles, IGF2, IGF1R, and IGF2R are only found in GCs. 90 However, some reports also found that human GCs and theca cells express the IGF1R. 88 , 122 Moreover, recent single‐cell RNA sequencing data showed that granulosa and theca cells express the IGF1R. 123 Thus, the relative distribution of IGFs and their receptor suggests that autocrine and paracrine IGF actions between GCs, the oocyte, and thecal cells are tightly coordinated during follicle maturation (Figure 5).

3.2.4. Bovine/swine/ovine

IGF2 and IGF1 have been detected in bovine GCs. 124 , 125 IGF1 protects GCs from apoptosis 126 and increases GC proliferation 127 , 128 via the IGF1R. In bovine GCs from small follicles, IGF1 acts synergistically with FSH to increase GC proliferation and CYP19A1 expression via the PI3K and AKT pathway. 129

IGF1 is produced by swine GCs 130 probably under the control of gonadotropins. 131 Accordingly, in the swine ovary, IGF1 and FSHR show selective follicular coexpression. 132 GCs also express IGF1R 133 and respond to IGFs with an increase in proliferation and steroid secretion. 133 , 134 , 135 IGFs synergize with FSH to increase steroid production, steroidogenic gene expression, and lipoprotein uptake 134 , 135 , 136 , 137 , 138 , 139 , 140 , 141 , 142 and promote FSH‐stimulated synthesis of hyaluronic acid and hyaluronan synthase 2. 143

In the ovine ovary, IGF1 is present in the GCs from secondary and antral follicles, 144 and its expression seems to be stimulated by GH or FSH. 145 IGF1 may also participate in cumulus expansion in response to FSH in vitro. 146 In addition, IGF1 and FSH synergistically increase oocyte growth and LH receptor expression in sheep follicle cultures. 144

4. IGF1R SIGNALING AND GC FUNCTION

This section outlines the effects that deletion or mutation of components of the IGF1R signaling pathway have on the regulation of GC function and female fertility.

4.1. IRS2

In rodents, targeted deletion of IRS1 does not affect fertility. 147 In marked contrast, deletion of IRS2 leads to female infertility and disturbances in glucose metabolism. 148 , 149 IRS2 KO female mice show reduced pituitary gonadotrophs and lower gonadotropin levels at proestrus; however, ovulation cannot be rescued by exogenous gonadotropins, 148 , 150 suggesting ovarian dysfunction. Thus, the reproductive defects observed in IRS2 KO mice confirm that IRS1 is insufficient to preserve ovarian function. 150 In Drosophila melanogaster, the deletion of chico, a homolog of IRS2, causes infertility. 151 , 152 Finally, ovarian IRS2 levels in humans are higher than those found in the liver or pancreas. 153 , 154 , 155 Thus, IRS2 seems crucial in regulating ovarian function, ranging from Drosophila to humans.

In preovulatory luteinizing GCs from FSH‐treated animals, FSH, cAMP, and IGF1 stimulate IRS2 expression 156 , 157 and phosphorylation. 158 Conversely, FSH stimulation of AKT, StAR/CYP11A1, and glucose uptake decreases in IRS2‐deficient GCs. 156 , 157 Strikingly, FSH weakly stimulates IRS2 expression in GCs from women with polycystic ovary syndrome (PCOS). 156 In mouse GCs, IRS2 expression closely relates to AKT activation. Thus, in vivo, treatment with an FSH analog increases IRS2 and AKT phosphorylation effects paralleled by a decrease in miR‐431. 159 MiR‐431 was previously identified as a regulator of IRS2 and IGF1R. 160 Conversely, miR‐431 overexpression decreases IRS2 expression and attenuates AKT activation. 159 In light of this evidence, FSH may control IGF1R signaling by regulating IRS2 activity via phosphorylation or miR‐431 expression.

4.2. SHP2

The role of phosphatases in the regulation of GCs remains understudied. Of particular interest are SHP1 and SHP2 (protein tyrosine phosphatase non‐receptor type 6 and type 11, respectively) since they can dephosphorylate IRSs. 161 , 162 SHP1 KO mice are markedly insulin‐sensitive because of enhanced IRS‐PI3K‐AKT signaling. 163 SHP1 and SHP2 are expressed in the ovary, 164 although SHP1 is present only in atretic follicles. 164 Mice lacking SHP2 in the GCs appear to have increased follicle growth and ovulation. 165 Thus, inhibition of SHP activity by FSH may enhance IGF1R signaling.

4.3. PI3K

The class IA PI3K enzymes are heterodimeric lipid kinases made up of a regulatory subunit, p85, and a catalytic subunit, of which three isoforms are known: p110α, p110β, and p110δ. 166 Of the catalytic subunits, p110δ (encoded by the Pik3cd gene) is highly expressed in the mouse ovary and plays a crucial role in the regulation of ovarian function. 167 Pik3cd KO females are subfertile, exhibit fewer growing follicles, have more atretic follicles, and respond poorly to exogenous gonadotropins. 167 Interestingly, primordial follicle activation and oocyte meiotic maturation are unaffected in Pik3cd KO mice. The findings indicate that the p110δ catalytic subunit is a critical component of the PI3K pathway and is needed for FSH stimulation of follicle growth.

4.4. PTEN

PTEN dephosphorylates PIP3 and inhibits PI3K signaling. Human, rat, and bovine GCs of large preovulatory follicles express higher levels of PTEN than GCs of small follicles, suggesting that PTEN expression increases during terminal follicular growth. 168 , 169 , 170 In human GCs, PTEN knockdown increases AKT phosphorylation and cell proliferation in response to IGF1. 171 Moreover, in human ovaries, there is an inverse relationship between PTEN and phospho‐AKT, confirming that PTEN antagonizes the activity of PI3K on AKT. 168 This was confirmed in mice lacking PTEN in GCs, which show increased ovulation, GC proliferation, and antral follicle formation. 172 Thus, the evidence indicates that PTEN controls AKT activation in GCs and promotes conditions favoring atresia/apoptosis of growing follicles. It is possible to foresee that an inhibitory effect of FSH on PTEN activity could enhance IGF1R signaling.

4.5. AKT

AKT activity is essential for the induction of GC differentiation by FSH. Thus, FSH actions are abolished by overexpression of dominant‐negative AKT 173 or treatment with AKT inhibitors. 31 It is also well‐known that FSH induces AKT phosphorylation in GCs via cAMP. 174 , 175 , 176 However, the mechanisms mediating the effects of cAMP on AKT remain controversial 177 since some reports showed that inhibition of PKA prevented AKT phosphorylation, 175 but others showed that PKA inhibition augments AKT phosphorylation by FSH. 11 , 70 , 176 , 178 As discussed below, FSH indirectly stimulates AKT activity by regulating the IGF1R signaling pathway.

Of the three isoforms, AKT1 and AKT2 are the major isoforms found in the ovary and GCs of rats and humans. 168 , 179 Genetic studies showed that AKT1KO mice are subfertile and display abnormal cyclicity, with ovaries containing fewer growing and preovulatory follicles refractory to exogenous gonadotropins. 180 At the molecular level, AKT1KO ovaries have decreased expression of the cell cycle regulators Ccnd3 and Ccnd1. 180 In contrast, fertility is not significantly affected in female mice lacking AKT2. 181 Thus, AKT1 is crucial for normal ovarian function and for the response of GCs to gonadotropins.

Recently, proteomic and genomic analyses of AKT1‐deficient GCs revealed defects in metabolism, apoptosis, cell cycle, and cytoskeleton dynamics. The report shows a decrease in the phosphorylation of proteins involved in intracellular transport and mitochondrial physiology, such as β‐catenin, RTN4, EEF1D, and CALM1, and an increase in phosphorylation of GOT2, EIF2S2, SRRM2, and PTMA, suggesting possible direct and indirect effects of AKT1 silencing on protein phosphorylation. 182 How these proteins regulate GC and ovarian function requires further investigation.

5. INTERDEPENDENT EFFECTS OF FSH AND IGFs IN GCs

As described thus far, IGFs have been considered to act as co‐gonadotropins functioning in an autocrine or paracrine manner to amplify FSH actions in the GCs. However, this view is not compatible with data showing that (i) FSH requires the presence of IGFs and IGF1R to stimulate GCs, (ii) FSH renders GCs sensitive to IGFs by supporting/controlling the IGF1R signaling pathway, and (iii) FSH signaling interacts with factors downstream of AKT to maximally stimulate GC differentiation.

  1. If IGFs only potentiate FSH actions, it is expected that blocking the availability of IGFs or IGF1R activity should partially but not completely block FSH stimulation of GC differentiation markers. In contrast, growing evidence shows that in the absence of IGFs, FSH is unable to stimulate GC differentiation. For instance, it was shown early on that blocking IGF availability with IGFBPs or anti‐IGF1 antibodies dose‐dependently and completely inhibits steroid and inhibin α production in rat 31 , 69 , 183 , 184 and human 112 , 113 GCs. IGFBPs also inhibit cAMP‐stimulated steroidogenesis, suggesting that the effects of blocking IGF1R activation are independent of FSHR activation. Moreover, the addition of IGF1 reverses the inhibitory effect of IGFBPs. 69 , 183 The crucial role of IGF1 on GC differentiation is further revealed by comparing the ovarian phenotypes observed in FSHKO, FSHRKO, and IGF1KO mice. FSHKO and FSHRKO mice are infertile due to a block in folliculogenesis at the preantral stage, 185 , 186 which is the same phenotype observed in IGF1KO and IGF1RGCKO mice, 56 , 73 , 74 demonstrating that in vivo IGF1 is also essential for the stimulation of antral follicle growth by FSH.

Similarly, a crucial role of IGF1R activation was demonstrated in experiments showing that FSH actions in GCs are inhibited by general tyrosine kinase inhibitors, 69 , 187 IGF1R‐specific inhibitors, or IGF1R knockdown. 30 , 31 Inhibition of IGF1R activity also completely blocks FSH‐induced phosphorylation of AKT in rodent and human GCs. 30 , 31 In vivo, FSH also fails to stimulate AKT in the absence of IGF actions, as shown in IGF1RGCKO mice. 56 Genomic‐wide analysis of human GCs treated with an IGF1R inhibitor revealed the full spectrum of genes regulated by IGFs and those regulated by FSH that required IGF1R activity. This report demonstrated that about 50% of FSH‐regulated genes require IGF1R activity and suggested that several aspects of GC function, including cholesterol synthesis, extracellular matrix production, reactive oxygen species metabolism, and cytoskeleton are coordinately regulated by FSH and IGFs in humans. 188

  • ii

    In rat GCs, IGF1 stimulates proliferation only in FSH‐primed cells, suggesting that pretreatment with FSH renders GCs responsive to IGF1. 189 One of the first pieces of evidence indicating that FSH enhances IGF1R signaling was their synergistic activation of AKT. 11 , 30 , 31 , 70 Thus, although IGF1 treatment of GCs maximally stimulated IGF1R tyrosine phosphorylation, it failed to stimulate AKT above FSH stimulatory levels. 30 , 31 Moreover, in GCs cultured in serum‐free media without FSH, AKT has minimal background stimulation even though GCs produce IGF1. 31 Since FSH treatment of rat or human GCs does not affect the phosphorylation of the IGF1R, 30 , 31 these findings suggest that FSH amplifies IGF1R signaling or removes inhibitory checkpoints located between the IGF1R and AKT.

  • iii

    Finally, it has been extensively demonstrated that although treatment of rodent GCs with IGF1 strongly activates AKT, 11 , 31 it does not stimulate the expression of genes involved in GC differentiation in the absence of FSH. 31 , 187 Moreover, the overexpression of constitutively active AKT (caAKT) alone is not enough to mimic the effects of FSH. 173 Hence, despite the presence of active AKT, GC differentiation fails in the absence of FSH, indicating a collaborative relationship between FSH signaling and AKT downstream targets in orchestrating GC differentiation. In agreement with this idea, the expression of caAKT is enough to amplify gene expression responses to FSH 30 , 31 , 173 and is further supported by findings demonstrating that in cells lacking IGF1R activity, FSH‐induced differentiation can be fully rescued by caAKT. 30

6. PLAUSIBLE MOLECULAR CROSSTALK BETWEEN FSH AND IGF INTRACELLULAR SIGNALING PATHWAYS

Evidence suggests that FSH and IGF1 signaling pathways intersect upstream and downstream of AKT to regulate gene expression and GC differentiation. In this section, we will integrate the above‐described findings and propose molecular mechanisms that may contribute to better understand the interaction between FSH and IGF signaling in GCs (Figure 6).

FIGURE 6.

FIGURE 6

Possible interactions between FSH and IGF1R signaling in GCs.

6.1. IGF1R to AKT

6.1.1. FSH inhibition of tyrosine phosphatases

Tyrosine phosphorylation of the IGF1R and IRS2 is essential for the formation of signaling complexes. Whether IGF1R and IRS2 interaction is affected by FSH remains to be determined. However, it has been shown that FSH increases IRS2 stability in GCs, 156 an effect that posttranslational modifications, including phosphorylation, could mediate. In fact, in the presence of FSH, higher levels of tyrosine‐phosphorylated IRS2 have been detected in GCs. 156 The mechanisms involved and whether IGF1R is required for IRS2 phosphorylation by FSH are currently not known. Since the FSHR does not directly regulate tyrosine kinases, it is most likely that FSH stimulates tyrosine phosphorylation of IRS2 by inhibiting tyrosine phosphatases such as SHP2, which have been shown to bind to and dephosphorylate IGF1R 190 , 191 , 192 and IRSs. 161 , 162

As described above, SHP2 negatively impacts GC function as SHP2 knockdown increases ovulation. 165 It is currently accepted that SHP1 and SHP2 are active under basal conditions, presumably by constitutive association with phosphoproteins, and that specific cell stimuli lead to their inactivation. SHP2 can be phosphorylated by Rous sarcoma oncogene kinase (SRC), protein kinase C (PKC), and PKA. 193 , 194 , 195 FSH targets each of these kinases 176 ; therefore, it may also regulate the activity of SHP2. Thus, in the absence of FSH, SHP2 dephosphorylates IRS2 blocking IGF1 signaling (Figure 6). This hypothesis infers that SHP2 participates in the mechanisms controlling IRS2 interaction with the IGF1R or PI3K and that FSH controls these interactions, options that remain unexplored.

PTEN silencing in GCs increases proliferation, antral follicle formation, and fertility. 172 Therefore, following the idea that FSH removes factors inhibiting IGF1R signaling, it is also possible that inhibition of PTEN activity by FSH amplifies IRS2 signals by removing its adverse effects on the PI3K pathway. However, the role of FSH in PTEN activity remains to be explored.

6.1.2. FSH inhibition of serine kinases

Although Tyr phosphorylation of IGF1R and IRS proteins is a key step in the activation of downstream signaling, IGF1R and IRS activity is also predetermined by serine/threonine phosphorylation levels. In contrast to the positive effect of tyrosine phosphorylation, serine/threonine phosphorylation of IGF1R and IRS has adverse effects on PI3K and AKT activation. Thus, an additional point of interaction between FSH and the IGF1R signaling pathway may be the phosphorylation of serine/threonine residues in IGF1R or IRS2 that prevent the activation of downstream signaling. For example, IRS phosphorylation on Ser residues by PKC, p70S6K, JNK, or GSK3β impairs IGF1R signaling and PI3K‐AKT activation. 196 , 197 , 198 , 199 , 200 , 201 , 202 , 203 , 204

The inhibitory effect of serine/threonine phosphorylation on IRS2 may explain the synergistic effect that H89 has on FSH‐induced AKT activation. 11 H89 is generally used as a PKA inhibitor; however, H89 also inhibits p70S6K. 205 Therefore, H89 may prevent p70S6K serine/threonine phosphorylation of IRS2 causing an increase in IGF1R signaling and potentiating the effect of FSH on AKT. Serine/threonine phosphorylation of IRS2 may also explain the inhibitory effect of PKC on FSH‐induced phosphorylation of AKT. 11

Evidence supports the role of serine phosphorylation in the regulation of IRS2 in GCs. For instance, FSH treatment of luteinized rat GCs decreases serine‐phosphorylated IRS2. 158 This effect could be mediated by serine/threonine phosphatases protein phosphatase 1 (PP1) and PP2A since okadaic acid and calyculin A, two inhibitors of PP1/PP2A, inhibit FSH‐stimulated steroidogenesis. 206 , 207 , 208 , 209 Accordingly, it has been shown that PP2A activity is stimulated by FSH and PKA. 210 , 211 Thus, FSH may contribute to PP1 and PP2A activation, which in turn dephosphorylate inhibitory serine residues on IRS2 or IGF1R stimulating their activity (Figure 6).

Finally, GSK3β has been shown to phosphorylate IRS at serine residues, an effect that attenuates downstream signaling. 203 , 204 GSK3β is a serine/threonine kinase inhibited via phosphorylation at serine 9. 212 In rat and porcine GCs, GSK3β phosphorylation at serine 9 increases after FSH treatment via a PI3K‐dependent mechanism. 213 , 214 , 215 It has been recently shown that a more robust phosphorylation at Ser9 occurs in GCs treated with both FSH and IGF1 and that inhibition of IGF1R activity prevents FSH phosphorylation of GSK3β. 215 These findings show that FSH and the IGF system interact to inactivate GSK3β. This idea is further supported by findings showing that the overexpression of constitutively active GSK3β blocks the synergistic stimulation of aromatase by FSH and IGF1, whereas inhibition of GSK3β activity potentiates the effects of FSH. 215 Therefore, it is possible to postulate that the strong inhibitory effects that FSH and IGF1 have on the activity of GSK3β may play an essential role in upregulating IRS2 activity (Figure 6).

6.2. AKT to target genes

Although AKT is essential, its activation alone is insufficient to stimulate GC differentiation, demonstrating that AKT interacts with FSH signaling independently of the IGF1R. AKT and FSH signaling interaction points include regulating CREB cofactors, inhibiting GSK3β/FOXO1, and eliminating PRAS40 activity (Figure 6). However, the full array of AKT downstream targets that could interact with the FSH‐activated signaling cascades warrants further investigation.

6.2.1. Regulation of CREB cofactors by AKT

Upon FSH activation, PKA phosphorylates CREB, enhancing its association with cofactors such as CBP and p300 216 , 217 and CREB‐regulated transcriptional coactivators (CRTCs), 218 which in turn modulate CREB activity. Of these cofactors, CRTCs appear to play a significant role in GCs. Indeed, CRTCs are highly expressed in GCs, and FSH stimulates CRTC2 nuclear localization, 219 , 220 contributing to activating gene expression. 221 On the other hand, CRTC activity is inhibited after phosphorylation by kinases such as salt‐inducible kinases (SIKs), which induce nuclear exclusion of CRTCs. 222 , 223 Therefore, factors that inhibit SIK may stimulate CRTCs and CREB. SIK activity increases upon phosphorylation by GSK3β 224 or decreases after phosphorylation by PKA. 225 As mentioned above, FSH inhibits GSK3β and stimulates PKA. Thus, CRTC/CREB activity may be controlled by a decrease in SIK activity following GSK3β inhibition or PKA stimulation (Figure 6).

In agreement with the negative effect of SIK activity on GC function, it was recently reported that SIKs participate in the regulation of folliculogenesis, ovulation, and fertility in rodents and the control of GC differentiation in humans and rodents. 226 Strikingly, it was shown that each SIK isoform has different effects on the regulation of female fertility and GC function. For instance, the knockdown of SIK2 enhances FSH‐induced GC differentiation and ovulation. In sharp contrast, the knockdown of SIK3 causes anovulation and female infertility. 226 Therefore, it is expected that if FSH regulates the activity of SIK kinases, this effect must be specific to SIK2. Further research is needed to determine this possibility.

6.2.2. FOXO1 inhibition

FOXO1 is highly expressed in GCs of growing follicles, 179 and its inhibition is a well‐known effect of AKT. 227 In rat GCs, FSH and IGF1 rapidly phosphorylate FOXO1 via activation of the PI3K/AKT signaling pathway, leading to its exclusion from the nucleus. 13 , 228 , 229 However, whether FSH and IGFs synergize to phosphorylate FOXO1 in GCs is not known. Mice lacking FOXO1 in the GCs are infertile, lack corpora lutea, and show follicles with abnormal appearances indicating that FOXO1 is essential for GC maturation and luteinization. At the molecular level, FOXO1 inhibition enhances the expression of the LH receptor, inhibin α, cyclin D2, epiregulin, LRH1, and several steroidogenesis genes. 228 , 229 , 230 This evidence suggests that inhibition of FOXO1 activity by AKT could be a crucial step needed to fully activate the differentiation program stimulated by FSH (Figure 6). Further research is needed to determine whether the detrimental effects of IGF1R or AKT inhibition could be rescued, at least partially, by the simultaneous knockdown of FOXO1.

6.2.3. PRAS40

The proline‐rich Akt substrate of 40 kDa (PRAS40, also known as AKT1S1) inhibits the mechanistic target of rapamycin (mTORC1). mTORC1 is a crucial regulator of protein synthesis known to be regulated by the IGF system via AKT. 231 In this pathway, AKT phosphorylates and inhibits PRAS40, 232 , 233 relieving mTORC1 inhibition. Inactivation of PRAS40 in Drosophila melanogaster increases egg laying and fertility. 234 Therefore, inhibition of PRAS40 by AKT could be a crucial step needed to stimulate protein synthesis by FSH and significantly enhance GC proliferation (Figure 6).

PRAS40 could also target mechanisms upstream of AKT. In Drosophila, inactivation of PRAS40 rescues the infertility of IRS2KO female flies (see above). 234 Moreover, in mice, the knockdown of PRAS40 causes a 4‐fold increase in IRS levels in hepatocytes, 235 indicating that PRAS40 inhibits IRS expression and could be a negative regulator of the IGF1R signaling in GCs. The capacity of PKA to phosphorylate PRAS40 236 supports the idea that FSH may also participate directly in the control of PRAS40 activity. Thus, PRAS40 may reduce IRS2 levels, thus inhibiting both the FSH and IGF1R pathways. This is an attractive hypothesis, but further experiments are needed to establish whether this occurs in GCs (Figure 6).

7. IGF1R AND FSHR CROSSTALK AND FEMALE INFERTILITY

Over 40% of female infertility cases stem from ovarian dysfunction due to a lack of oocyte maturation or due to anovulation. 237 Addressing this challenge involves weeks of hormone treatments, particularly the administration of exogenous FSH, aimed at fostering the maturation of multiple ovarian follicles to maximize the collection of mature oocytes for in vitro fertilization. Regrettably, up to 24% of women exhibit poor responses to FSH stimulation, yielding very few mature oocytes for collection. 238 This insufficiency in egg production accounts for over 80% of canceled IVF cycles. 239 The mechanisms underlying FSH‐induced follicle maturation in the human ovary remain poorly understood, partly due to the absence of a robust model for studying human GC differentiation, further complicating these investigations.

Intraovarian IGF system inefficiencies correlate with decreased ovarian function in women. Thus, components of the IGF gene family are downregulated in GCs of women with diminished ovarian reserve. 240 Clinical data support close interactions between FSH and IGFs in follicle development by showing that IGF levels are low in the follicular fluid of poor‐responding patients. 241 On the other hand, IGF concentrations in follicular fluid are higher in women who respond better to follicular stimulation. 242 In addition, serum IGF1 levels decline with age, which correlates with the well‐known decrease in fertility due to aging. 243 These findings underscore the close interaction between FSH and IGFs in regulating human follicle development.

Defects in IGF1R signaling have also been implicated in the development of PCOS, which is characterized by follicle growth disorders, chronic anovulation, hyperandrogenemia, and polycystic ovaries. Thus, PCOS is associated with AKT and GSK3β polymorphisms downstream of IGF1R. 244 Defective expression of the IGF1R in GCs and increased IGF1R expression in theca cells are common features in PCOS ovaries. 245 This imbalance may be responsible, at least in part, for impaired insulin and IGF1 actions in GCs and enhanced actions in thecal stromal cells in PCOS ovaries leading to hyperandrogenism. 122 IGFR1 expression in preantral follicles, particularly in early preantral PCOS follicles, suggests a role for IGFs in regulating the initiation of human follicle growth. Thus, accelerated preantral follicle growth in PCOS may result from increased endogenous IGF activity. 245 Therefore, understanding IGF1R signaling pathways may help elucidate GC and theca cell defects driving follicle growth changes in PCOS.

8. CONCLUSIONS

We propose that ovarian follicles exposed to increasing levels of FSH benefit from locally produced IGFs, and only follicles with high endogenous IGF production can fully respond to FSH. Thus, a positive feedback loop between the endocrine effects of FSH and the autocrine and paracrine actions of IGFs plays an essential role in the establishment of follicle dominance and follicle survival in all species. Understanding the complexity of the molecular and signaling pathways coordinating the actions of FSH and IGFs in the GCs could reveal novel therapeutic targets to optimize follicle growth in subfertile patients. These targets can also be used to develop novel contraceptive approaches.

FUNDING INFORMATION

This work was supported by the National Institutes of Health grant number R01HD097202. The funding source had no role in the design, practice, or analysis of this study.

CONFLICT OF INTEREST STATEMENT

The authors declare no conflict of interest could be perceived as prejudicing the impartiality of the research reported.

ETHICS STATEMENT

Human/Animal rights: This review article contains no studies of human and animal subjects directly performed by authors. There is also no clinical trial involved.

Hayes E, Winston N, Stocco C. Molecular crosstalk between insulin‐like growth factors and follicle‐stimulating hormone in the regulation of granulosa cell function. Reprod Med Biol. 2024;23:e12575. 10.1002/rmb2.12575

REFERENCES

  • 1. Neirijnck Y, Papaioannou MD, Nef S. The insulin/IGF system in mammalian sexual development and reproduction. Int J Mol Sci. 2019;20(18):4440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ipsa E, Cruzat VF, Kagize JN, Yovich JL, Keane KN. Growth hormone and insulin‐like growth factor action in reproductive tissues. Front Endocrinol. 2019;10:777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gougeon A. Dynamics of follicular growth in the human: a model from preliminary results. Hum Reprod. 1986;1(2):81–87. [DOI] [PubMed] [Google Scholar]
  • 4. George JW, Dille EA, Heckert LL. Current concepts of follicle‐stimulating hormone receptor gene regulation. Biol Reprod. 2011;84(1):7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Carlone DL, Richards JS. Evidence that functional interactions of CREB and SF‐1 mediate hormone regulated expression of the aromatase gene in granulosa cells and constitutive expression in R2C cells. J Steroid Biochem Mol Biol. 1997;61(3–6):223–231. [PubMed] [Google Scholar]
  • 6. Carlone DL, Richards JS. Functional interactions, phosphorylation, and levels of 3′, 5′‐cyclic adenosine monophosphate‐regulatory element binding protein and steroidogenic factor‐1 mediate hormone‐regulated and constitutive expression of aromatase in gonadal cells. Mol Endocrinol. 1997;11(3):292–304. [DOI] [PubMed] [Google Scholar]
  • 7. Stocco C, Kwintkiewicz J, Cai Z. Identification of regulatory elements in the Cyp19 proximal promoter in rat luteal cells. J Mol Endocrinol. 2007;39(4):211–221. [DOI] [PubMed] [Google Scholar]
  • 8. Kwintkiewicz J, Cai Z, Stocco C. Follicle‐stimulating hormone‐induced activation of Gata4 contributes in the up‐regulation of Cyp19 expression in rat granulosa cells. Mol Endocrinol. 2007;21(4):933–947. [DOI] [PubMed] [Google Scholar]
  • 9. Cottom J, Salvador LM, Maizels ET, Reierstad S, Park Y, Carr DW, et al. Follicle‐stimulating hormone activates extracellular signal‐regulated kinase but not extracellular signal‐regulated kinase kinase through a 100‐kDa phosphotyrosine phosphatase. J Biol Chem. 2003;278(9):7167–7179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Maizels ET, Cottom J, Jones JC, Hunzicker‐Dunn M. Follicle stimulating hormone (FSH) activates the p38 mitogen‐activated protein kinase pathway, inducing small heat shock protein phosphorylation and cell rounding in immature rat ovarian granulosa cells. Endocrinology. 1998;139(7):3353–3356. [DOI] [PubMed] [Google Scholar]
  • 11. Gonzalez IJ, Falender AE, Ochsner S, Firestone GL, Richards JS. Follicle‐stimulating hormone (FSH) stimulates phosphorylation and activation of protein kinase B (PKB/Akt) and serum and glucocorticoid‐lnduced kinase (Sgk): evidence for a kinase‐independent signaling by FSH in granulosa cells. Mol Endocrinol. 2000;14(8):1283–1300. [DOI] [PubMed] [Google Scholar]
  • 12. Matsuda F, Inoue N, Maeda A, Cheng Y, Sai T, Gonda H, et al. Expression and function of apoptosis initiator FOXO3 in granulosa cells during follicular atresia in pig ovaries. J Reprod Dev. 2011;57(1):151–158. [DOI] [PubMed] [Google Scholar]
  • 13. Shen M, Liu Z, Li B, Teng Y, Zhang J, Tang Y, et al. Involvement of FoxO1 in the effects of follicle‐stimulating hormone on inhibition of apoptosis in mouse granulosa cells. Cell Death Dis. 2014;5:e1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Alam H, Weck J, Maizels E, Park Y, Lee EJ, Ashcroft M, et al. Role of the phosphatidylinositol‐3‐kinase and extracellular regulated kinase pathways in the induction of hypoxia‐inducible factor (HIF)‐1 activity and the HIF‐1 target vascular endothelial growth factor in ovarian granulosa cells in response to follicle‐stimulating hormone. Endocrinology. 2009;150(2):915–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Casarini L, Crépieux P. Molecular mechanisms of action of FSH. Front Endocrinol. 2019;10:305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ulloa‐Aguirre A, Reiter E, Crepieux P. FSH receptor signaling: complexity of interactions and signal diversity. Endocrinology. 2018;159(8):3020–3035. [DOI] [PubMed] [Google Scholar]
  • 17. Casarini L, Simoni M. Recent advances in understanding gonadotropin signaling. Fac Rev. 2021;10:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chauvin S, Cohen‐Tannoudji J, Guigon CJ. Estradiol signaling at the heart of Folliculogenesis: its potential deregulation in human ovarian pathologies. Int J Mol Sci. 2022;23(1):512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fisch B, Rose MP, Elder MG, Winston RM, Margara RA, Hillier SG. Effects of oestrogen on progesterone synthesis and arachidonic acid metabolism in human luteal cells. Clin Endocrinol (Oxf). 1994;40(1):21–32. [DOI] [PubMed] [Google Scholar]
  • 20. Stocco C. Aromatase expression in the ovary: hormonal and molecular regulation. Steroids. 2008;73(5):473–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Chia DJ. Minireview: mechanisms of growth hormone‐mediated gene regulation. Mol Endocrinol. 2014;28(7):1012–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Daughaday WH, Rotwein P. Insulin‐like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr Rev. 1989;10(1):68–91. [DOI] [PubMed] [Google Scholar]
  • 23. Livingstone C, Borai A. Insulin‐like growth factor‐II: its role in metabolic and endocrine disease. Clin Endocrinol (Oxf). 2014;80(6):773–781. [DOI] [PubMed] [Google Scholar]
  • 24. Siddle K. Molecular basis of signaling specificity of insulin and IGF receptors: neglected corners and recent advances. Front Endocrinol. 2012;3:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Antonetti DA, Algenstaedt P, Kahn CR. Insulin receptor substrate 1 binds two novel splice variants of the regulatory subunit of phosphatidylinositol 3‐kinase in muscle and brain. Mol Cell Biol. 1996;16(5):2195–2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pearce LR, Komander D, Alessi DR. The nuts and bolts of AGC protein kinases. Nat Rev Mol Cell Biol. 2010;11(1):9–22. [DOI] [PubMed] [Google Scholar]
  • 27. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell. 2007;129(7):1261–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Girnita L, Worrall C, Takahashi S, Seregard S, Girnita A. Something old, something new and something borrowed: emerging paradigm of insulin‐like growth factor type 1 receptor (IGF‐1R) signaling regulation. Cell Mol Life Sci. 2014;71(13):2403–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Baumgarten SC, Convissar SM, Zamah AM, Fierro MA, Winston NJ, Scoccia B, et al. FSH regulates IGF‐2 expression in human granulosa cells in an AKT‐dependent manner. J Clin Endocrinol Metab. 2015;100(8):E1046–E1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Baumgarten SC, Convissar SM, Fierro MA, Winston NJ, Scoccia B, Stocco C. IGF1R signaling is necessary for fsh‐induced activation of AKT and differentiation of human cumulus granulosa cells. J Clin Endocrinol Metab. 2014;99(8):2995–3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Zhou P, Baumgarten SC, Wu Y, Bennett J, Winston N, Hirshfeld‐Cytron J, et al. IGF‐I signaling is essential for FSH stimulation of AKT and steroidogenic genes in granulosa cells. Mol Endocrinol. 2013;27(3):511–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Schmidt B, Kiecke‐Siemsen C, Waheed A, Braulke T, von Figura K. Localization of the insulin‐like growth factor II binding site to amino acids 1508‐1566 in repeat 11 of the mannose 6‐phosphate/insulin‐like growth factor II receptor. J Biol Chem. 1995;270(25):14975–14982. [DOI] [PubMed] [Google Scholar]
  • 33. Xu Y, Papageorgiou A, Polychronakos C. Developmental regulation of the soluble form of insulin‐like growth factor‐II/mannose 6‐phosphate receptor in human serum and amniotic fluid. J Clin Endocrinol Metab. 1998;83(2):437–442. [DOI] [PubMed] [Google Scholar]
  • 34. Mazerbourg S, Monget P. Insulin‐like growth factor binding proteins and IGFBP proteases: a dynamic system regulating the ovarian Folliculogenesis. Front Endocrinol. 2018;9:134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Spitschak M, Hoeflich A. Potential functions of IGFBP‐2 for ovarian Folliculogenesis and steroidogenesis. Front Endocrinol. 2018;9:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Murphy LJ, Bell GI, Friesen HG. Tissue distribution of insulin‐like growth factor‐I and factor‐ii messenger‐ribonucleic‐acid in the adult‐rat. Endocrinology. 1987;120(4):1279–1282. [DOI] [PubMed] [Google Scholar]
  • 37. Oliver JE, Aitman TJ, Powell JF, Wilson CA, Clayton RN. Insulin‐like growth factor I gene expression in the rat ovary is confined to the granulosa cells of developing follicles. Endocrinology. 1989;124(6):2671–2679. [DOI] [PubMed] [Google Scholar]
  • 38. Zhou J, Chin E, Bondy C. Cellular pattern of insulin‐like growth factor‐I (IGF‐I) and IGF‐I receptor gene expression in the developing and mature ovarian follicle. Endocrinology. 1991;129(6):3281–3288. [DOI] [PubMed] [Google Scholar]
  • 39. Zhou J, Refuerzo J, Bondy C. Granulosa‐cell DNA‐synthesis is strictly correlated with the presence of insulin‐like growth‐factor‐I and absence of C‐Fos/C‐Jun expression. Mol Endocrinol. 1995;9(7):924–931. [DOI] [PubMed] [Google Scholar]
  • 40. Hernandez ER, Roberts CT Jr, LeRoith D, Adashi EY. Rat ovarian insulin‐like growth factor I (IGF‐I) gene expression is granulosa cell‐selective: 5′‐untranslated mRNA variant representation and hormonal regulation. Endocrinology. 1989;125(1):572–574. [DOI] [PubMed] [Google Scholar]
  • 41. Botero LF, Roberts CT, Leroith D, Adashi EY, Hernandez ER. Insulin‐like growth factor‐I gene‐expression by primary cultures of ovarian‐cells ‐ insulin and dexamethasone dependence. Endocrinology. 1993;132(6):2703–2708. [DOI] [PubMed] [Google Scholar]
  • 42. Sugino N, Telleria CM, Tessier C, Gibori G. Regulation and role of the insulin‐like growth factor I system in rat luteal cells. J Reprod Fertil. 1999;115(2):349–355. [DOI] [PubMed] [Google Scholar]
  • 43. Parmer TG, Roberts CT Jr, LeRoith D, Adashi EY, Khan I, Solan N, et al. Expression, action, and steroidal regulation of insulin‐like growth factor‐I (IGF‐I) and IGF‐I receptor in the rat corpus luteum: their differential role in the two cell populations forming the corpus luteum. Endocrinology. 1991;129(6):2924–2932. [DOI] [PubMed] [Google Scholar]
  • 44. Davoren JB, Kasson BG, Li CH, Hsueh AJW. Specific insulin‐like growth‐factor (Igf) I‐binding and ii‐binding sites on rat granulosa‐cells—relation to Igf action. Endocrinology. 1986;119(5):2155–2162. [DOI] [PubMed] [Google Scholar]
  • 45. Adashi EY, Resnick CE, Svoboda ME, Vanwyk JJ. Follicle‐stimulating‐hormone enhances somatomedin‐C binding to cultured rat granulosa‐cells ‐ evidence for camp dependence. J Biol Chem. 1986;261(9):3923–3926. [PubMed] [Google Scholar]
  • 46. Adashi EY, Resnick CE, Hernandez ER, Svoboda ME, Vanwyk JJ. Characterization and regulation of a specific cell‐membrane receptor for somatomedin‐C insulin‐like growth factor‐I in cultured rat granulosa‐cells. Endocrinology. 1988;122(1):194–201. [DOI] [PubMed] [Google Scholar]
  • 47. Hernandez ER, Hurwitz A, Botero L, Ricciarelli E, Werner H, Roberts CT Jr, et al. Insulin‐like growth factor receptor gene expression in the rat ovary: divergent regulation of distinct receptor species. Mol Endocrinol. 1991;5(12):1799–1805. [DOI] [PubMed] [Google Scholar]
  • 48. Hernandez ER, Resnick CE, Svoboda ME, Vanwyk JJ, Payne DW, Adashi EY. Somatomedin‐C insulin‐like growth factor‐I as an enhancer of androgen biosynthesis by cultured rat ovarian‐cells. Endocrinology. 1988;122(4):1603–1612. [DOI] [PubMed] [Google Scholar]
  • 49. Cara JF, Rosenfield RL. Insulin‐like growth factor I and insulin potentiate luteinizing hormone‐induced androgen synthesis by rat ovarian thecal‐interstitial cells. Endocrinology. 1988;123(2):733–739. [DOI] [PubMed] [Google Scholar]
  • 50. Damario MA, Bogovich K, Liu HC, Rosenwaks Z, Poretsky L. Synergistic effects of insulin‐like growth factor‐I and human chorionic gonadotropin in the rat ovary. Metabolism. 2000;49(3):314–320. [DOI] [PubMed] [Google Scholar]
  • 51. Magoffin DA, Kurtz KM, Erickson GF. Insulin‐like growth factor‐I selectively stimulates cholesterol side‐chain cleavage expression in ovarian theca‐interstitial cells. Mol Endocrinol. 1990;4(3):489–496. [DOI] [PubMed] [Google Scholar]
  • 52. Magoffin DA, Weitsman SR. Insulin‐like growth factor‐I stimulates the expression of 3 beta‐hydroxysteroid dehydrogenase messenger ribonucleic acid in ovarian theca‐interstitial cells. Biol Reprod. 1993;48(5):1166–1173. [DOI] [PubMed] [Google Scholar]
  • 53. Magoffin DA, Weitsman SR. Effect of insulin‐like growth factor‐I on cholesterol side‐chain cleavage cytochrome P450 messenger ribonucleic acid expression in ovarian theca‐interstitial cells stimulated to differentiate in vitro. Mol Cell Endocrinol. 1993;96(1–2):45–51. [DOI] [PubMed] [Google Scholar]
  • 54. Magoffin DA, Weitsman SR. Differentiation of ovarian theca‐interstitial cells in vitro: regulation of 17 alpha‐hydroxylase messenger ribonucleic acid expression by luteinizing hormone and insulin‐like growth factor‐I. Endocrinology. 1993;132(5):1945–1951. [DOI] [PubMed] [Google Scholar]
  • 55. Duleba AJ, Spaczynski RZ, Olive DL, Behrman HR. Effects of insulin and insulin‐like growth factors on proliferation of rat ovarian theca‐interstitial cells. Biol Reprod. 1997;56(4):891–897. [DOI] [PubMed] [Google Scholar]
  • 56. Baumgarten SC, Armouti M, Ko C, Stocco C. IGF1R expression in ovarian granulosa cells is essential for steroidogenesis, follicle survival, and fertility in female mice. Endocrinology. 2017;158(7):2309–2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Pitetti JL, Torre D, Conne B, Papaioannou MD, Cederroth CR, Xuan S, et al. Insulin receptor and IGF1R are not required for oocyte growth, differentiation, and maturation in mice. Sex Dev. 2009;3(5):264–272. [DOI] [PubMed] [Google Scholar]
  • 58. Adashi EY, Resnick CE, Svoboda ME, Van Wyk JJ. A novel role for somatomedin‐C in the cytodifferentiation of the ovarian granulosa cell. Endocrinology. 1984;115(3):1227–1229. [DOI] [PubMed] [Google Scholar]
  • 59. Adashi EY, Resnick CE, Svoboda ME, Van Wyk JJ. Somatomedin‐C synergizes with follicle‐stimulating hormone in the acquisition of progestin biosynthetic capacity by cultured rat granulosa cells. Endocrinology. 1985;116(6):2135–2142. [DOI] [PubMed] [Google Scholar]
  • 60. Davoren JB, Hsueh JW, Li CH. Somatomedin C augments FSH‐induced differentiation of cultured rat granulosa cells. Am J Physiol. 1985;249(1 Pt 1):E26–E33. [DOI] [PubMed] [Google Scholar]
  • 61. Davoren JB, Hsueh AJW. Insulin enhances Fsh‐stimulated steroidogenesis by cultured rat granulosa‐cells. Mol Cell Endocrinol. 1984;35(2–3):97–105. [DOI] [PubMed] [Google Scholar]
  • 62. Bley MA, Simon JC, Estevez AG, de Asua LJ, Baranao JL. Effect of follicle‐stimulating hormone on insulin‐like growth factor‐I‐stimulated rat granulosa cell deoxyribonucleic acid synthesis. Endocrinology. 1992;131(3):1223–1229. [DOI] [PubMed] [Google Scholar]
  • 63. Adashi EY, Resnick CE, Svoboda ME, Van Wyk JJ. Somatomedin‐C enhances induction of luteinizing hormone receptors by follicle‐stimulating hormone in cultured rat granulosa cells. Endocrinology. 1985;116(6):2369–2375. [DOI] [PubMed] [Google Scholar]
  • 64. Adashi EY, Resnick CE, Brodie AM, Svoboda ME, Van Wyk JJ. Somatomedin‐C‐mediated potentiation of follicle‐stimulating hormone‐induced aromatase activity of cultured rat granulosa cells. Endocrinology. 1985;117(6):2313–2320. [DOI] [PubMed] [Google Scholar]
  • 65. Adashi EY, Resnick CE, Svoboda ME, Van Wyk JJ. Somatomedin‐C as an amplifier of follicle‐stimulating‐hormone action ‐ enhanced accumulation of Adenosine‐3′,5′‐monophosphate. Endocrinology. 1986;118(1):149–155. [DOI] [PubMed] [Google Scholar]
  • 66. Adashi EY, Resnick CE, Svoboda ME, Vanwyk JJ, Hascall VC, Yanagishita M. Independent and synergistic actions of somatomedin‐C in the stimulation of proteoglycan biosynthesis by cultured rat granulosa‐cells. Endocrinology. 1986;118(1):456–458. [DOI] [PubMed] [Google Scholar]
  • 67. deMoura MD, Choi D, Adashi EY, Payne DW. Insulin‐like growth factor‐I‐mediated amplification of follicle‐stimulating hormone‐supported progesterone accumulation by cultured rat granulosa cells: enhancement of steroidogenic enzyme activity and expression. Biol Reprod. 1997;56(4):946–953. [DOI] [PubMed] [Google Scholar]
  • 68. Zhang ZW, Carson RS, Herington AC, Lee VWK, Burger HG. Follicle‐stimulating‐hormone and somatomedin‐C stimulate inhibin production by rat granulosa‐cells Invitro. Endocrinology. 1987;120(4):1633–1638. [DOI] [PubMed] [Google Scholar]
  • 69. Li D, Kubo T, Kim H, Shimasaki S, Erickson GF. Endogenous insulin‐like growth factor‐I is obligatory for stimulation of rat inhibin alpha‐subunit expression by follicle‐stimulating hormone. Biol Reprod. 1998;58(1):219–225. [DOI] [PubMed] [Google Scholar]
  • 70. Sun GW, Kobayashi H, Suzuki M, Kanayama N, Terao T. Follicle‐stimulating hormone and insulin‐like growth factor I synergistically induce up‐regulation of cartilage link protein (Crtl1) via activation of phosphatidylinositol‐dependent kinase/Akt in rat granulosa cells. Endocrinology. 2003;144(3):793–801. [DOI] [PubMed] [Google Scholar]
  • 71. Hirakawa T, Minegishi T, Abe K, Kishi H, Ibuki Y, Miyamoto K. A role of insulin‐like growth factor I in luteinizing hormone receptor expression in granulosa cells. Endocrinology. 1999;140(11):4965–4971. [DOI] [PubMed] [Google Scholar]
  • 72. Minegishi T, Hirakawa T, Kishi H, Abe K, Abe Y, Mizutani T, et al. A role of insulin‐like growth factor I for follicle‐stimulating hormone receptor expression in rat granulosa cells. Biol Reprod. 2000;62(2):325–333. [DOI] [PubMed] [Google Scholar]
  • 73. Baker J, Hardy MP, Zhou J, Bondy C, Lupu F, Bellve AR, et al. Effects of an Igf1 gene null mutation on mouse reproduction. Mol Endocrinol. 1996;10(7):903–918. [DOI] [PubMed] [Google Scholar]
  • 74. Zhou J, Kumar TR, Matzuk MM, Bondy C. Insulin‐like growth factor I regulates gonadotropin responsiveness in the murine ovary. Mol Endocrinol. 1997;11(13):1924–1933. [DOI] [PubMed] [Google Scholar]
  • 75. Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying null mutations of the genes encoding insulin‐like growth factor I (Igf‐1) and type 1 IGF receptor (Igf1r). Cell. 1993;75(1):59–72. [PubMed] [Google Scholar]
  • 76. Baker J, Liu JP, Robertson EJ, Efstratiadis A. Role of insulin‐like growth‐factors in embryonic and postnatal‐growth. Cell. 1993;75(1):73–82. [PubMed] [Google Scholar]
  • 77. DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of the mouse insulin‐like growth factor II gene. Cell. 1991;64(4):849–859. [DOI] [PubMed] [Google Scholar]
  • 78. Zhou J, Bievre M, Bondy CA. Reduced GLUT1 expression in IGF1 null oocytes and follicles. Growth Horm IGF Res. 2000;10(3):111–117. [DOI] [PubMed] [Google Scholar]
  • 79. Kadakia R, Arraztoa JA, Bondy C, Zhou J. Granulosa cell proliferation is impaired in the Igf1 null ovary. Growth Horm IGF Res. 2001;11(4):220–224. [DOI] [PubMed] [Google Scholar]
  • 80. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, et al. IGF‐1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature. 2003;421(6919):182–187. [DOI] [PubMed] [Google Scholar]
  • 81. Holzenberger M, Hamard G, Zaoui R, Leneuve P, Ducos B, Beccavin C, et al. Experimental IGF‐I receptor deficiency generates a sexually dimorphic pattern of organ‐specific growth deficits in mice, affecting fat tissue in particular. Endocrinology. 2001;142(10):4469–4478. [DOI] [PubMed] [Google Scholar]
  • 82. Ludwig T, Eggenschwiler J, Fisher P, D'Ercole AJ, Davenport ML, Efstratiadis A. Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null backgrounds. Dev Biol. 1996;177(2):517–535. [DOI] [PubMed] [Google Scholar]
  • 83. Sekulovski N, Whorton AE, Shi M, Hayashi K, MacLean JA 2nd. Periovulatory insulin signaling is essential for ovulation, granulosa cell differentiation, and female fertility. FASEB J. 2020;34(2):2376–2391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bachelot A, Monget P, Imbert‐Bollore P, Coshigano K, Kopchick JJ, Kelly PA, et al. Growth hormone is required for ovarian follicular growth. Endocrinology. 2002;143(10):4104–4112. [DOI] [PubMed] [Google Scholar]
  • 85. Zaczek D, Hammond J, Suen L, Wandji S, Service D, Bartke A, et al. Impact of growth hormone resistance on female reproductive function: new insights from growth hormone receptor knockout mice. Biol Reprod. 2002;67(4):1115–1124. [DOI] [PubMed] [Google Scholar]
  • 86. Slot KA, Kastelijn J, Bachelot A, Kelly PA, Binart N, Teerds KJ. Reduced recruitment and survival of primordial and growing follicles in GH receptor‐deficient mice. Reproduction. 2006;131(3):525–532. [DOI] [PubMed] [Google Scholar]
  • 87. Stratikopoulos E, Szabolcs M, Dragatsis I, Klinakis A, Efstratiadis A. The hormonal action of IGF1 in postnatal mouse growth. Proc Natl Acad Sci U S A. 2008;105(49):19378–19383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Man L, Lustgarten Guahmich N, Kallinos E, Park L, Caiazza B, Khan M, et al. Exogenous insulin‐like growth factor 1 accelerates growth and maturation of follicles in human cortical xenografts and increases ovarian output in mice. F S Sci. 2021;2(3):237–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Geisthovel F, Moretti‐Rojas I, Asch RH, Rojas FJ. Expression of insulin‐like growth factor‐II (IGF‐II) messenger ribonucleic acid (mRNA), but not IGF‐I mRNA, in human preovulatory granulosa cells. Hum Reprod. 1989;4(8):899–902. [DOI] [PubMed] [Google Scholar]
  • 90. el‐Roeiy A, Chen X, Roberts VJ, LeRoith D, Roberts CT, Yen SS. Expression of insulin‐like growth factor‐I (IGF‐I) and IGF‐II and the IGF‐I, IGF‐II, and insulin receptor genes and localization of the gene products in the human ovary. J Clin Endocrinol Metab. 1993;77(5):1411–1418. [DOI] [PubMed] [Google Scholar]
  • 91. Qu J, Godin PA, Nisolle M, Donnez J. Expression of receptors for insulin‐like growth factor‐I and transforming growth factor‐beta in human follicles. Mol Hum Reprod. 2000;6(2):137–145. [DOI] [PubMed] [Google Scholar]
  • 92. Voutilainen R, Franks S, Mason HD, Martikainen H. Expression of insulin‐like growth factor (IGF), IGF‐binding protein, and IGF receptor messenger ribonucleic acids in normal and polycystic ovaries. J Clin Endocrinol Metab. 1996;81(3):1003–1008. [DOI] [PubMed] [Google Scholar]
  • 93. Klein NA, Battaglia DE, Woodruff TK, Padmanabhan V, Giudice LC, Bremner WJ, et al. Ovarian follicular concentrations of activin, follistatin, inhibin, insulin‐like growth factor I (IGF‐I), IGF‐II, IGF‐binding protein‐2 (IGFBP‐2), IGFBP‐3, and vascular endothelial growth factor in spontaneous menstrual cycles of normal women of advanced reproductive age. J Clin Endocrinol Metab. 2000;85(12):4520–4525. [DOI] [PubMed] [Google Scholar]
  • 94. Seifer DB, Giudice LC, Dsupin BA, Haning RV Jr, Frishman GN, Burger HG. Follicular fluid insulin‐like growth factor‐I and insulin‐like growth factor‐II concentrations vary as a function of day 3 serum follicle stimulating hormone. Hum Reprod. 1995;10(4):804–806. [DOI] [PubMed] [Google Scholar]
  • 95. Kaya A, Atabekoglu CS, Kahraman K, Taskin S, Ozmen B, Berker B, et al. Follicular fluid concentrations of IGF‐I, IGF‐II, IGFBP‐3, VEGF, AMH, and inhibin‐B in women undergoing controlled ovarian hyperstimulation using GnRH agonist or GnRH antagonist. Eur J Obstet Gynecol Reprod Biol. 2012;164(2):167–171. [DOI] [PubMed] [Google Scholar]
  • 96. Thierry van Dessel HJ, Chandrasekher Y, Yap OW, Lee PD, Hintz RL, Faessen GH, et al. Serum and follicular fluid levels of insulin‐like growth factor I (IGF‐ I), IGF‐II, and IGF‐binding protein‐1 and ‐3 during the normal menstrual cycle. J Clin Endocrinol Metab. 1996;81(3):1224–1231. [DOI] [PubMed] [Google Scholar]
  • 97. Wang TH, Chang CL, Wu HM, Chiu YM, Chen CK, Wang HS. Insulin‐like growth factor‐II (IGF‐II), IGF‐binding protein‐3 (IGFBP‐3), and IGFBP‐4 in follicular fluid are associated with oocyte maturation and embryo development. Fertil Steril. 2006;86(5):1392–1401. [DOI] [PubMed] [Google Scholar]
  • 98. Penarrubia J, Balasch J, Garcia‐Bermudez M, Casamitjana R, Vanrell JA, Hernandez ER. Growth hormone does not increase the expression of insulin‐like growth factors and their receptor genes in the pre‐menopausal human ovary. Hum Reprod. 2000;15(6):1241–1246. [DOI] [PubMed] [Google Scholar]
  • 99. Dor J, Ben‐Shlomo I, Lunenfeld B, Pariente C, Levran D, Karasik A, et al. Insulin‐like growth factor‐I (IGF‐I) may not be essential for ovarian follicular development: evidence from IGF‐I deficiency. J Clin Endocrinol Metab. 1992;74(3):539–542. [DOI] [PubMed] [Google Scholar]
  • 100. Menashe Y, Sack J, Mashiach S. Spontaneous pregnancies in two women with Laron‐type dwarfism: are growth hormone and circulating insulin‐like growth factor mandatory for induction of ovulation? Hum Reprod. 1991;6(5):670–671. [DOI] [PubMed] [Google Scholar]
  • 101. Zeleznik AJ, Little‐Ihrig L, Ramasawamy S. Administration of insulin‐like growth factor I to rhesus monkeys does not augment gonadotropin‐stimulated ovarian steroidogenesis. J Clin Endocrinol Metab. 2002;87(12):5722–5729. [DOI] [PubMed] [Google Scholar]
  • 102. Devoto L, Christenson LK, McAllister JM, Makrigiannakis A, Strauss JF 3rd. Insulin and insulin‐like growth factor‐I and ‐II modulate human granulosa‐lutein cell steroidogenesis: enhancement of steroidogenic acute regulatory protein (StAR) expression. Mol Hum Reprod. 1999;5(11):1003–1010. [DOI] [PubMed] [Google Scholar]
  • 103. Kamada S, Kubota T, Taguchi M, Ho WR, Sakamoto S, Aso T. Effects of insulin‐like growth factor‐II on proliferation and differentiation of ovarian granulosa cells. Horm Res. 1992;37(4–5):141–149. [DOI] [PubMed] [Google Scholar]
  • 104. Mason HD, Willis DS, Holly JM, Franks S. Insulin preincubation enhances insulin‐like growth factor‐II (IGF‐II) action on steroidogenesis in human granulosa cells. J Clin Endocrinol Metab. 1994;78(5):1265–1267. [DOI] [PubMed] [Google Scholar]
  • 105. Kubota T, Kamada S, Ohara M, Taguchi M, Sakamoto S, Shimizu Y, et al. Insulin‐like growth factor II in follicular fluid of the patients with in vitro fertilization and embryo transfer. Fertil Steril. 1993;59(4):844–849. [DOI] [PubMed] [Google Scholar]
  • 106. Steinkampf MP, Mendelson CR, Simpson ER. Effects of epidermal growth factor and insulin‐like growth factor I on the levels of mRNA encoding aromatase cytochrome P‐450 of human ovarian granulosa cells. Mol Cell Endocrinol. 1988;59(1–2):93–99. [DOI] [PubMed] [Google Scholar]
  • 107. Erickson GF, Garzo VG, Magoffin DA. Insulin‐like growth factor‐I regulates aromatase activity in human granulosa and granulosa luteal cells. J Clin Endocrinol Metab. 1989;69(4):716–724. [DOI] [PubMed] [Google Scholar]
  • 108. Yong EL, Baird DT, Yates R, Reichert LE Jr, Hillier SG. Hormonal regulation of the growth and steroidogenic function of human granulosa cells. J Clin Endocrinol Metab. 1992;74(4):842–849. [DOI] [PubMed] [Google Scholar]
  • 109. Bergh C, Olsson JH, Hillensjo T. Effect of insulin‐like growth factor I on steroidogenesis in cultured human granulosa cells. Acta Endocrinol. 1991;125(2):177–185. [DOI] [PubMed] [Google Scholar]
  • 110. Di Blasio AM, Vigano P, Ferrari A. Insulin‐like growth factor‐II stimulates human granulosa‐luteal cell proliferation in vitro. Fertil Steril. 1994;61(3):483–487. [DOI] [PubMed] [Google Scholar]
  • 111. Martinez JC, Stouffer RL, Hazzard TM, Patton PE, Molskness TA. Insulin‐like growth factors‐1 and ‐2, but not hypoxia, synergize with gonadotropin hormone to promote vascular endothelial growth factor‐a secretion by monkey granulosa cells from preovulatory follicles. Biol Reprod. 2003;68(4):1112–1118. [DOI] [PubMed] [Google Scholar]
  • 112. Yuan W, Giudice LC. Insulin‐like growth factor‐II mediates the steroidogenic and growth promoting actions of follicle stimulating hormone on human ovarian pre‐antral follicles cultured in vitro. J Clin Endocrinol Metab. 1999;84(4):1479–1482. [DOI] [PubMed] [Google Scholar]
  • 113. Willis DS, Mason HD, Watson H, Franks S. Developmentally regulated responses of human granulosa cells to insulin‐like growth factors (IGFs): IGF‐I and IGF‐II action mediated via the type‐I IGF receptor. J Clin Endocrinol Metab. 1998;83(4):1256–1259. [DOI] [PubMed] [Google Scholar]
  • 114. Tkachenko OY, Wolf S, Lawson MS, Ting AY, Rodrigues JK, Xu F, et al. Insulin‐like growth factor 2 is produced by antral follicles and promotes preantral follicle development in macaquesdagger. Biol Reprod. 2021;104(3):602–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Louhio H, Hovatta O, Sjoberg J, Tuuri T. The effects of insulin, and insulin‐like growth factors I and II on human ovarian follicles in long‐term culture. Mol Hum Reprod. 2000;6(8):694–698. [DOI] [PubMed] [Google Scholar]
  • 116. Poretsky L, Chun B, Liu HC, Rosenwaks Z. Insulin‐like growth factor II (IGF‐II) inhibits insulin‐like growth factor binding protein I (IGFBP‐1) production in luteinized human granulosa cells with a potency similar to insulin‐like growth factor I (IGF‐I) and higher than insulin. J Clin Endocrinol Metab. 1996;81(9):3412–3414. [DOI] [PubMed] [Google Scholar]
  • 117. Erickson GF, Magoffin DA, Cragun JR, Chang RJ. The effects of insulin and insulin‐like growth factors‐I and ‐II on estradiol production by granulosa cells of polycystic ovaries. J Clin Endocrinol Metab. 1990;70(4):894–902. [DOI] [PubMed] [Google Scholar]
  • 118. Voutilainen R, Miller WL. Coordinate tropic hormone regulation of mRNAs for insulin‐like growth factor II and the cholesterol side‐chain‐cleavage enzyme, P450scc [corrected], in human steroidogenic tissues. Proc Natl Acad Sci U S A. 1987;84(6):1590–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Ramasharma K, Li CH. Human pituitary and placental hormones control human insulin‐like growth factor II secretion in human granulosa cells. Proc Natl Acad Sci U S A. 1987;84(9):2643–2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Hobeika E, Armouti M, Kala H, Fierro MA, Winston NJ, Scoccia B, et al. Oocyte‐secreted factors synergize with FSH to promote aromatase expression in primary human cumulus cells. J Clin Endocrinol Metab. 2019;104(5):1667–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Hobeika E, Armouti M, Fierro MA, Winston N, Scoccia H, Zamah AM, et al. Regulation of insulin‐like growth factor 2 by oocyte‐secreted factors in primary human granulosa cells. J Clin Endocrinol Metab. 2020;105(1):327–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Samoto T, Maruo T, Matsuo H, Katayama K, Barnea ER, Mochizuki M. Altered expression of insulin and insulin‐like growth factor‐I receptors in follicular and stromal compartments of polycystic ovaries. Endocr J. 1993;40(4):413–424. [DOI] [PubMed] [Google Scholar]
  • 123. Man L, Lustgarten‐Guahmich N, Kallinos E, Redhead‐Laconte Z, Liu S, Schattman B, et al. Comparison of human antral follicles of xenograft versus ovarian origin reveals disparate molecular signatures. Cell Rep. 2020;32(6):108027. [DOI] [PubMed] [Google Scholar]
  • 124. Schams D, Berisha B, Kosmann M, Amselgruber W. Expression and localization of IGF family members in bovine antral follicles during final growth and in luteal tissue during different stages of estrous cycle and pregnancy. Domest Anim Endocrinol. 2002;22(1):51–72. [DOI] [PubMed] [Google Scholar]
  • 125. Armstrong DG, Gutierrez CG, Baxter G, Glazyrin AL, Mann GE, Woad KJ, et al. Expression of mRNA encoding IGF‐I, IGF‐II and type 1 IGF receptor in bovine ovarian follicles. J Endocrinol. 2000;165(1):101–113. [DOI] [PubMed] [Google Scholar]
  • 126. Hu CL, Cowan RG, Harman RM, Quirk SM. Cell cycle progression and activation of Akt kinase are required for insulin‐like growth factor I‐mediated suppression of apoptosis in granulosa cells. Mol Endocrinol. 2004;18(2):326–338. [DOI] [PubMed] [Google Scholar]
  • 127. Savion N, Lui GM, Laherty R, Gospodarowicz D. Factors controlling proliferation and progesterone production by bovine granulosa cells in serum‐free medium. Endocrinology. 1981;109(2):409–420. [DOI] [PubMed] [Google Scholar]
  • 128. Armstrong DT, Xia P, deGannes G, Tekpetey FR, Khamsi F. Differential effects of insulin‐like growth factor‐I and follicle‐stimulating hormone on proliferation and differentiation of bovine cumulus cells and granulosa cells. Biol Reprod. 1996;54(2):331–338. [DOI] [PubMed] [Google Scholar]
  • 129. Mani AM, Fenwick MA, Cheng Z, Sharma MK, Singh D, Wathes DC. IGF1 induces up‐regulation of steroidogenic and apoptotic regulatory genes via activation of phosphatidylinositol‐dependent kinase/AKT in bovine granulosa cells. Reproduction. 2010;139(1):139–151. [DOI] [PubMed] [Google Scholar]
  • 130. Hammond JM, Knight AP, Rechler MM. Somatomedin secretion by porcine granulosa‐cells—a potential mechanism for regulating ovarian follicular‐growth. Clin Res. 1984;32(2):A485‐A. [Google Scholar]
  • 131. Hammond JM, Hsu CJ, Klindt J, Tsang BK, Downey BR. Gonadotropins increase concentrations of Immunoreactive insulin‐like growth factor‐I in porcine follicular‐fluid Invivo. Biol Reprod. 1988;38(2):304–308. [DOI] [PubMed] [Google Scholar]
  • 132. Zhou J, Adesanya OO, Vatzias G, Hammond JM, Bondy CA. Selective expression of insulin‐like growth factor system components during porcine ovary follicular selection. Endocrinology. 1996;137(11):4893–4901. [DOI] [PubMed] [Google Scholar]
  • 133. Baranao JLS, Hammond JM. Comparative effects of insulin and insulin‐like growth‐factors on DNA‐synthesis and differentiation of porcine granulosa‐cells. Biochem Biophys Res Commun. 1984;124(2):484–490. [DOI] [PubMed] [Google Scholar]
  • 134. Veldhuis JD, Demers LM. A role for somatomedin‐C as a differentiating hormone and amplifier of hormone action on ovarian‐cells—studies with synthetically pure human somatomedin‐C and swine granulosa‐cells. Biochem Biophys Res Commun. 1985;130(1):234–240. [DOI] [PubMed] [Google Scholar]
  • 135. Garmey JC, Day RN, Day KH, Veldhuis JD. Mechanisms of regulation of ovarian sterol‐metabolism by insulin‐like growth‐factor type‐ii ‐ in‐vitro studies with swine granulosa‐cells. Endocrinology. 1993;133(2):800–808. [DOI] [PubMed] [Google Scholar]
  • 136. Veldhuis JD, Rodgers RJ, Dee A, Simpson ER. The insulin‐like growth‐factor, somatomedin‐C, induces the synthesis of cholesterol side‐chain cleavage cytochrome‐P‐450 and Adrenodoxin in ovarian‐cells. J Biol Chem. 1986;261(6):2499–2502. [PubMed] [Google Scholar]
  • 137. Veldhuis JD, Rodgers RJ, Furlanetto RW, Azimi P, Juchter D, Garmey J. Synergistic actions of estradiol and the insulin‐like growth‐factor somatomedin‐C on swine ovarian (granulosa) cells. Endocrinology. 1986;119(2):530–538. [DOI] [PubMed] [Google Scholar]
  • 138. Balasubramanian K, Lavoie HA, Garmey JC, Stocco DM, Veldhuis JD. Regulation of porcine granulosa cell steroidogenic acute regulatory protein (StAR) by insulin‐like growth factor I: synergism with follicle‐stimulating hormone or protein kinase a agonist. Endocrinology. 1997;138(1):433–439. [DOI] [PubMed] [Google Scholar]
  • 139. LaVoie HA, Garmey JC, Day RN, Veldhuis JD. Concerted regulation of low density lipoprotein receptor gene expression by follicle‐stimulating hormone and insulin‐like growth factor I in porcine granulosa cells: promoter activation, messenger ribonucleic acid stability, and sterol feedback. Endocrinology. 1999;140(1):178–186. [DOI] [PubMed] [Google Scholar]
  • 140. LaVoie HA, Benoit AM, Garmey JC, Dailey RA, Wright DJ, Veldhuis JD. Coordinate developmental expression of genes regulating sterol economy and cholesterol side‐chain cleavage in the porcine ovary. Biol Reprod. 1997;57(2):402–407. [DOI] [PubMed] [Google Scholar]
  • 141. Veldhuis JD, Rodgers RJ. Mechanisms subserving the steroidogenic synergism between follicle‐stimulating‐hormone and insulin‐like growth factor‐I (somatomedin‐C)—alterations in cellular sterol‐metabolism in swine granulosa‐cells. J Biol Chem. 1987;262(16):7658–7664. [PubMed] [Google Scholar]
  • 142. LaVoie HA, Garmey JC, Veldhuis JD. Mechanisms of insulin‐like growth factor I augmentation of follicle‐stimulating hormone‐induced porcine steroidogenic acute regulatory protein gene promoter activity in granulosa cells. Endocrinology. 1999;140(1):146–153. [DOI] [PubMed] [Google Scholar]
  • 143. Nemcova L, Nagyova E, Petlach M, Tomanek M, Prochazka R. Molecular mechanisms of insulin‐like growth factor 1 promoted synthesis and retention of hyaluronic acid in porcine oocyte‐cumulus complexes. Biol Reprod. 2007;76(6):1016–1024. [DOI] [PubMed] [Google Scholar]
  • 144. Monte APO, Barros VRP, Santos JM, Menezes VG, Cavalcante AYP, Gouveia BB, et al. Immunohistochemical localization of insulin‐like growth factor‐1 (IGF‐1) in the sheep ovary and the synergistic effect of IGF‐1 and FSH on follicular development in vitro and LH receptor immunostaining. Theriogenology. 2019;129:61–69. [DOI] [PubMed] [Google Scholar]
  • 145. Khalid M, Haresign W, Luck M. Secretion of IGF‐1 by ovine granulosa cells: effects of growth hormone and follicle stimulating hormone. Anim Reprod Sci. 2000;58(3–4):261–272. [DOI] [PubMed] [Google Scholar]
  • 146. Singh B, Armstrong DT. Insulin‐like growth factor‐1, a component of serum that enables porcine cumulus cells to expand in response to follicle‐stimulating hormone in vitro. Biol Reprod. 1997;56(6):1370–1375. [DOI] [PubMed] [Google Scholar]
  • 147. Araki E, Lipes MA, Patti ME, Bruning JC, Haag B 3rd, Johnson RS, et al. Alternative pathway of insulin signalling in mice with targeted disruption of the IRS‐1 gene. Nature. 1994;372(6502):186–190. [DOI] [PubMed] [Google Scholar]
  • 148. Burks DJ, Font de Mora J, Schubert M, Withers DJ, Myers MG, Towery HH, et al. IRS‐2 pathways integrate female reproduction and energy homeostasis. Nature. 2000;407(6802):377–382. [DOI] [PubMed] [Google Scholar]
  • 149. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S, et al. Disruption of IRS‐2 causes type 2 diabetes in mice. Nature. 1998;391(6670):900–904. [DOI] [PubMed] [Google Scholar]
  • 150. Neganova I, Al‐Qassab H, Heffron H, Selman C, Choudhury AI, Lingard SJ, et al. Role of central nervous system and ovarian insulin receptor substrate 2 signaling in female reproductive function in the mouse. Biol Reprod. 2007;76(6):1045–1053. [DOI] [PubMed] [Google Scholar]
  • 151. Bohni R, Riesgo‐Escovar J, Oldham S, Brogiolo W, Stocker H, Andruss BF, et al. Autonomous control of cell and organ size by CHICO, a drosophila homolog of vertebrate IRS1‐4. Cell. 1999;97(7):865–875. [DOI] [PubMed] [Google Scholar]
  • 152. Tatar M, Kopelman A, Epstein D, Tu MP, Yin CM, Garofalo RS. A mutant drosophila insulin receptor homolog that extends life‐span and impairs neuroendocrine function. Science. 2001;292(5514):107–110. [DOI] [PubMed] [Google Scholar]
  • 153. Yen HW, Jakimiuk AJ, Munir I, Magoffin DA. Selective alterations in insulin receptor substrates‐1, −2 and −4 in theca but not granulosa cells from polycystic ovaries. Mol Hum Reprod. 2004;10(7):473–479. [DOI] [PubMed] [Google Scholar]
  • 154. Wu X, Sallinen K, Anttila L, Makinen M, Luo C, Pollanen P, et al. Expression of insulin‐receptor substrate‐1 and ‐2 in ovaries from women with insulin resistance and from controls. Fertil Steril. 2000;74(3):564–572. [DOI] [PubMed] [Google Scholar]
  • 155. Vassen L, Wegrzyn W, Klein‐Hitpass L. Human insulin receptor substrate‐2 (IRS‐2) is a primary progesterone response gene. Mol Endocrinol. 1999;13(3):485–494. [DOI] [PubMed] [Google Scholar]
  • 156. Anjali G, Kaur S, Lakra R, Taneja J, Kalsey GS, Nagendra A, et al. FSH stimulates IRS‐2 expression in human granulosa cells through cAMP/SP1, an inoperative FSH action in PCOS patients. Cell Signal. 2015;27(12):2452–2466. [DOI] [PubMed] [Google Scholar]
  • 157. Lei L, Han F, Cui Q, Liao W, Liu H, Guan G, et al. IRS2 depletion inhibits cell proliferation and decreases hormone secretion in mouse granulosa cells. J Reprod Dev. 2018;64(5):409–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Chahal N, Geethadevi A, Kaur S, Lakra R, Nagendra A, Shrivastav TG, et al. Direct impact of gonadotropins on glucose uptake and storage in preovulatory granulosa cells: implications in the pathogenesis of polycystic ovary syndrome. Metabolism. 2021;115:154458. [DOI] [PubMed] [Google Scholar]
  • 159. Yang L, Lv Q, Liu J, Qi S, Fu D. miR‐431 regulates granulosa cell function through the IRS2/PI3K/AKT signaling pathway. J Reprod Dev. 2020;66(3):231–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Tanaka T, Sugaya S, Kita K, Arai M, Kanda T, Fujii K, et al. Inhibition of cell viability by human IFN‐beta is mediated by microRNA‐431. Int J Oncol. 2012;40(5):1470–1476. [DOI] [PubMed] [Google Scholar]
  • 161. Myers MG Jr, Mendez R, Shi P, Pierce JH, Rhoads R, White MF. The COOH‐terminal tyrosine phosphorylation sites on IRS‐1 bind SHP‐2 and negatively regulate insulin signaling. J Biol Chem. 1998;273(41):26908–26914. [DOI] [PubMed] [Google Scholar]
  • 162. Ling Y, Maile LA, Clemmons DR. Tyrosine phosphorylation of the beta3‐subunit of the alphaVbeta3 integrin is required for embrane association of the tyrosine phosphatase SHP‐2 and its further recruitment to the insulin‐like growth factor I receptor. Mol Endocrinol. 2003;17(9):1824–1833. [DOI] [PubMed] [Google Scholar]
  • 163. Dubois MJ, Bergeron S, Kim HJ, Dombrowski L, Perreault M, Fournes B, et al. The SHP‐1 protein tyrosine phosphatase negatively modulates glucose homeostasis. Nat Med. 2006;12(5):549–556. [DOI] [PubMed] [Google Scholar]
  • 164. Russell DL, Richards JS. Differentiation‐dependent prolactin responsiveness and stat (signal transducers and activators of transcription) signaling in rat ovarian cells. Mol Endocrinol. 1999;13(12):2049–2064. [DOI] [PubMed] [Google Scholar]
  • 165. Wei X, Zheng L, Tian Y, Wang H, Su Y, Feng G, et al. Tyrosine phosphatase SHP2 in ovarian granulosa cells balances follicular development by inhibiting PI3K/AKT signaling. J Mol Cell Biol. 2022;14(7):mjac048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3‐kinase pathway in cancer. Nat Rev Drug Discov. 2009;8(8):627–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Li Q, He H, Zhang YL, Li XM, Guo X, Huo R, et al. Phosphoinositide 3‐kinase p110delta mediates estrogen‐ and FSH‐stimulated ovarian follicle growth. Mol Endocrinol. 2013;27(9):1468–1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Goto M, Iwase A, Ando H, Kurotsuchi S, Harata T, Kikkawa F. PTEN and Akt expression during growth of human ovarian follicles. J Assist Reprod Genet. 2007;24(11):541–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Froment P, Bontoux M, Pisselet C, Monget P, Dupont J. PTEN expression in ovine granulosa cells increases during terminal follicular growth. FEBS Lett. 2005;579(11):2376–2382. [DOI] [PubMed] [Google Scholar]
  • 170. Namli Kalem M, Anadol E, Kalem Z, Sezginer PY, Elmas C, Yilmaz C, et al. A rat study on the PTEN expression in ovarian tissue in PCOS and folliculogenesis. Sci Rep. 2023;13(1):20774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Goto M, Iwase A, Harata T, Takigawa S, Suzuki K, Manabe S, et al. IGF1‐induced AKT phosphorylation and cell proliferation are suppressed with the increase in PTEN during luteinization in human granulosa cells. Reproduction. 2009;137(5):835–842. [DOI] [PubMed] [Google Scholar]
  • 172. Fan HY, Liu Z, Cahill N, Richards JS. Targeted disruption of Pten in ovarian granulosa cells enhances ovulation and extends the life span of luteal cells. Mol Endocrinol. 2008;22(9):2128–2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Zeleznik AJ, Saxena D, Little‐Ihrig L. Protein kinase B is obligatory for follicle‐stimulating hormone‐induced granulosa cell differentiation. Endocrinology. 2003;144(9):3985–3994. [DOI] [PubMed] [Google Scholar]
  • 174. Richards JS, Russell DL, Ochsner S, Hsieh M, Doyle KH, Falender AE, et al. Novel signaling pathways that control ovarian follicular development, ovulation, and luteinization. Recent Prog Horm Res. 2002;57:195–220. [DOI] [PubMed] [Google Scholar]
  • 175. Hunzicker‐Dunn ME, Lopez‐Biladeau B, Law NC, Fiedler SE, Carr DW, Maizels ET. PKA and GAB2 play central roles in the FSH signaling pathway to PI3K and AKT in ovarian granulosa cells. Proc Natl Acad Sci U S A. 2012;109(44):E2979–E2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Wayne CM, Fan HY, Cheng X, Richards JS. Follicle‐stimulating hormone induces multiple signaling cascades: evidence that activation of Rous sarcoma oncogene, RAS, and the epidermal growth factor receptor are critical for granulosa cell differentiation. Mol Endocrinol. 2007;21(8):1940–1957. [DOI] [PubMed] [Google Scholar]
  • 177. Stocco C. Follicle‐stimulating hormone (FSH) phosphorylation of protein kinase B (AKT) remains controversial. J Biol Chem. 2016;291(27):14385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Chen Y‐J, Hsiao P‐W, Lee M‐T, Mason JI, Ke F‐C, Hwang J‐J. Interplay of PI3K and cAMP/PKA signaling, and rapamycin‐hypersensitivity in TGFβ1 enhancement of FSH‐stimulated steroidogenesis in rat ovarian granulosa cells. J Endocrinol. 2007;192(2):405–419. [DOI] [PubMed] [Google Scholar]
  • 179. Richards JS, Sharma SC, Falender AE, Lo YH. Expression of FKHR, FKHRL1, and AFX genes in the rodent ovary: evidence for regulation by IGF‐I, estrogen, and the gonadotropins. Mol Endocrinol. 2002;16(3):580–599. [DOI] [PubMed] [Google Scholar]
  • 180. Brown C, LaRocca J, Pietruska J, Ota M, Anderson L, Smith SD, et al. Subfertility caused by altered follicular development and oocyte growth in female mice lacking PKB alpha/Akt1. Biol Reprod. 2010;82(2):246–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Restuccia DF, Hynx D, Hemmings BA. Loss of PKBbeta/Akt2 predisposes mice to ovarian cyst formation and increases the severity of polycystic ovary formation in vivo. Dis Model Mech. 2012;5(3):403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Elzaiat M, Herman L, Legois B, Leger T, Todeschini AL, Veitia RA. High‐throughput exploration of the network dependent on AKT1 in mouse ovarian granulosa cells. Mol Cell Proteomics. 2019;18(7):1307–1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Bicsak TA, Shimonaka M, Malkowski M, Ling N. Insulin‐like growth factor‐binding protein (IGF‐BP) inhibition of granulosa cell function: effect on cyclic adenosine 3′,5′‐monophosphate, deoxyribonucleic acid synthesis, and comparison with the effect of an IGF‐I antibody. Endocrinology. 1990;126(4):2184–2189. [DOI] [PubMed] [Google Scholar]
  • 184. Adashi EY, Resnick CE, Payne DW, Rosenfeld RG, Matsumoto T, Hunter MK, et al. The mouse intraovarian insulin‐like growth factor I system: departures from the rat paradigm. Endocrinology. 1997;138(9):3881–3890. [DOI] [PubMed] [Google Scholar]
  • 185. Dierich A, Sairam MR, Monaco L, Fimia GM, Gansmuller A, LeMeur M, et al. Impairing follicle‐stimulating hormone (FSH) signaling in vivo: targeted disruption of the FSH receptor leads to aberrant gametogenesis and hormonal imbalance. Proc Natl Acad Sci U S A. 1998;95(23):13612–13617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Kumar TR, Wang Y, Lu N, Matzuk MM. Follicle stimulating hormone is required for ovarian follicle maturation but not male fertility. Nat Genet. 1997;15(2):201–204. [DOI] [PubMed] [Google Scholar]
  • 187. Eimerl S, Orly J. Regulation of steroidogenic genes by insulin‐like growth factor‐1 and follicle‐stimulating hormone: differential responses of cytochrome P450 side‐chain cleavage, steroidogenic acute regulatory protein, and 3beta‐hydroxysteroid dehydrogenase/isomerase in rat granulosa cells. Biol Reprod. 2002;67(3):900–910. [DOI] [PubMed] [Google Scholar]
  • 188. Stocco C, Baumgarten SC, Armouti M, Fierro MA, Winston NJ, Scoccia B, et al. Genome‐wide interactions between FSH and insulin‐like growth factors in the regulation of human granulosa cell differentiation. Hum Reprod. 2017;32(4):905–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Kanzaki M, Hattori M, Kojima I. Growth or differentiation: determination by FSH of the action of insulin‐like growth factor‐I in cultured rat granulosa cells. Endocr J. 1996;43(1):15–23. [DOI] [PubMed] [Google Scholar]
  • 190. Kenner KA, Anyanwu E, Olefsky JM, Kusari J. Protein‐tyrosine phosphatase 1B is a negative regulator of insulin‐ and insulin‐like growth factor‐I‐stimulated signaling. J Biol Chem. 1996;271(33):19810–19816. [DOI] [PubMed] [Google Scholar]
  • 191. Maile LA, Clemmons DR. The alphaVbeta3 integrin regulates insulin‐like growth factor I (IGF‐I) receptor phosphorylation by altering the rate of recruitment of the Src‐homology 2‐containing phosphotyrosine phosphatase‐2 to the activated IGF‐I receptor. Endocrinology. 2002;143(11):4259–4264. [DOI] [PubMed] [Google Scholar]
  • 192. Maile LA, Clemmons DR. Regulation of insulin‐like growth factor I receptor dephosphorylation by SHPS‐1 and the tyrosine phosphatase SHP‐2. J Biol Chem. 2002;277(11):8955–8960. [DOI] [PubMed] [Google Scholar]
  • 193. Kawakami T, Xiao W, Yasudo H, Kawakami Y. Regulation of proliferation, survival, differentiation, and activation by the signaling platform for SHP‐1 phosphatase. Adv Biol Regul. 2012;52(1):7–15. [DOI] [PubMed] [Google Scholar]
  • 194. Liu Y, Kruhlak MJ, Hao JJ, Shaw S. Rapid T cell receptor‐mediated SHP‐1 S591 phosphorylation regulates SHP‐1 cellular localization and phosphatase activity. J Leukoc Biol. 2007;82(3):742–751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Burmeister BT, Taglieri DM, Wang L, Carnegie GK. Src homology 2 domain‐containing phosphatase 2 (Shp2) is a component of the A‐kinase‐anchoring protein (AKAP)‐Lbc complex and is inhibited by protein kinase a (PKA) under pathological hypertrophic conditions in the heart. J Biol Chem. 2012;287(48):40535–40546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, et al. Protein kinase C theta inhibits insulin signaling by phosphorylating IRS1 at Ser(1101). J Biol Chem. 2004;279(44):45304–45307. [DOI] [PubMed] [Google Scholar]
  • 197. Tremblay F, Brule S, Hee Um S, Li Y, Masuda K, Roden M, et al. Identification of IRS‐1 Ser‐1101 as a target of S6K1 in nutrient‐ and obesity‐induced insulin resistance. Proc Natl Acad Sci U S A. 2007;104(35):14056–14061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Ravichandran LV, Esposito DL, Chen J, Quon MJ. Protein kinase C‐zeta phosphorylates insulin receptor substrate‐1 and impairs its ability to activate phosphatidylinositol 3‐kinase in response to insulin. J Biol Chem. 2001;276(5):3543–3549. [DOI] [PubMed] [Google Scholar]
  • 199. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c‐Jun NH(2)‐terminal kinase promotes insulin resistance during association with insulin receptor substrate‐1 and phosphorylation of Ser(307). J Biol Chem. 2000;275(12):9047–9054. [DOI] [PubMed] [Google Scholar]
  • 200. Weigert C, Hennige AM, Brischmann T, Beck A, Moeschel K, Schauble M, et al. The phosphorylation of Ser318 of insulin receptor substrate 1 is not per se inhibitory in skeletal muscle cells but is necessary to trigger the attenuation of the insulin‐stimulated signal. J Biol Chem. 2005;280(45):37393–37399. [DOI] [PubMed] [Google Scholar]
  • 201. Moeschel K, Beck A, Weigert C, Lammers R, Kalbacher H, Voelter W, et al. Protein kinase C‐zeta‐induced phosphorylation of Ser318 in insulin receptor substrate‐1 (IRS‐1) attenuates the interaction with the insulin receptor and the tyrosine phosphorylation of IRS‐1. J Biol Chem. 2004;279(24):25157–25163. [DOI] [PubMed] [Google Scholar]
  • 202. Hennige AM, Stefan N, Kapp K, Lehmann R, Weigert C, Beck A, et al. Leptin down‐regulates insulin action through phosphorylation of serine‐318 in insulin receptor substrate 1. FASEB J. 2006;20(8):1206–1208. [DOI] [PubMed] [Google Scholar]
  • 203. Eldar‐Finkelman H, Krebs EG. Phosphorylation of insulin receptor substrate 1 by glycogen synthase kinase 3 impairs insulin action. Proc Natl Acad Sci U S A. 1997;94(18):9660–9664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Liberman Z, Eldar‐Finkelman H. Serine 332 phosphorylation of insulin receptor substrate‐1 by glycogen synthase kinase‐3 attenuates insulin signaling. J Biol Chem. 2005;280(6):4422–4428. [DOI] [PubMed] [Google Scholar]
  • 205. Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351(Pt 1):95–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Azhar S, Wang H, Tsai L, Reaven E. Okadaic acid interferes with lipoprotein‐supported corticosterone production in adrenal cells. Biochem Biophys Res Commun. 1991;179(2):726–733. [DOI] [PubMed] [Google Scholar]
  • 207. Azhar S, Frazier JA, Tsai L, Reaven E. Effect of okadaic acid on utilization of lipoprotein‐derived cholesteryl esters by rat steroidogenic cells. J Lipid Res. 1994;35(7):1161–1176. [PubMed] [Google Scholar]
  • 208. Sewer MB, Waterman MR. Adrenocorticotropin/cyclic adenosine 3′,5′‐monophosphate‐mediated transcription of the human CYP17 gene in the adrenal cortex is dependent on phosphatase activity. Endocrinology. 2002;143(5):1769–1777. [DOI] [PubMed] [Google Scholar]
  • 209. Sewer MB, Waterman MR. cAMP‐dependent transcription of steroidogenic genes in the human adrenal cortex requires a dual‐specificity phosphatase in addition to protein kinase a. J Mol Endocrinol. 2002;29(1):163–174. [DOI] [PubMed] [Google Scholar]
  • 210. Levallet G, Levallet J, Bonnamy PJ. FSH‐induced phosphoprotein phosphatase 2A‐mediated deactivation of particulate phosphodiesterase‐4 activities is abolished after alteration in proteoglycan synthesis in immature rat Sertoli cells. J Endocrinol. 2008;197(1):45–54. [DOI] [PubMed] [Google Scholar]
  • 211. Feschenko MS, Stevenson E, Nairn AC, Sweadner KJ. A novel cAMP‐stimulated pathway in protein phosphatase 2A activation. J Pharmacol Exp Ther. 2002;302(1):111–118. [DOI] [PubMed] [Google Scholar]
  • 212. Dajani R, Fraser E, Roe SM, Young N, Good V, Dale TC, et al. Crystal structure of glycogen synthase kinase 3 beta: structural basis for phosphate‐primed substrate specificity and autoinhibition. Cell. 2001;105(6):721–732. [DOI] [PubMed] [Google Scholar]
  • 213. Alliston TN, Gonzalez‐Robayna IJ, Buse P, Firestone GL, Richards JS. Expression and localization of serum/glucocorticoid‐induced kinase in the rat ovary: relation to follicular growth and differentiation. Endocrinology. 2000;141(1):385–395. [DOI] [PubMed] [Google Scholar]
  • 214. Gillio‐Meina C, Hui YY, LaVoie HA. Expression of CCAAT/enhancer binding proteins alpha and beta in the porcine ovary and regulation in primary cultures of granulosa cells. Biol Reprod. 2005;72(5):1194–1204. [DOI] [PubMed] [Google Scholar]
  • 215. Convissar S, Bennett‐Toomey J, Stocco C. Insulin‐like growth factor 1 enhances follicle‐stimulating hormone‐induced phosphorylation of GATA4 in rat granulosa cells. Mol Cell Endocrinol. 2023;559:111807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Lundblad JR, Kwok RP, Laurance ME, Harter ML, Goodman RH. Adenoviral E1A‐associated protein p300 as a functional homologue of the transcriptional co‐activator CBP. Nature. 1995;374(6517):85–88. [DOI] [PubMed] [Google Scholar]
  • 217. Parker D, Ferreri K, Nakajima T, LaMorte VJ, Evans R, Koerber SC, et al. Phosphorylation of CREB at Ser‐133 induces complex formation with CREB‐binding protein via a direct mechanism. Mol Cell Biol. 1996;16(2):694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Conkright MD, Canettieri G, Screaton R, Guzman E, Miraglia L, Hogenesch JB, et al. TORCs: transducers of regulated CREB activity. Mol Cell. 2003;12(2):413–423. [DOI] [PubMed] [Google Scholar]
  • 219. Armouti M, Rodriguez‐Esquivel M, Stocco C. Mechanism of negative modulation of FSH signaling by salt‐inducible kinases in rat granulosa cells. Front Endocrinol. 2022;13:1026358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Fang WL, Lee MT, Wu LS, Chen YJ, Mason J, Ke FC, et al. CREB coactivator CRTC2/TORC2 and its regulator calcineurin crucially mediate follicle‐stimulating hormone and transforming growth factor beta1 upregulation of steroidogenesis. J Cell Physiol. 2012;227(6):2430–2440. [DOI] [PubMed] [Google Scholar]
  • 221. Altarejos JY, Montminy M. CREB and the CRTC co‐activators: sensors for hormonal and metabolic signals. Nat Rev Mol Cell Biol. 2011;12(3):141–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Katoh Y, Takemori H, Lin XZ, Tamura M, Muraoka M, Satoh T, et al. Silencing the constitutive active transcription factor CREB by the LKB1‐SIK signaling cascade. FEBS J. 2006;273(12):2730–2748. [DOI] [PubMed] [Google Scholar]
  • 223. Screaton RA, Conkright MD, Katoh Y, Best JL, Canettieri G, Jeffries S, et al. The CREB coactivator TORC2 functions as a calcium‐ and cAMP‐sensitive coincidence detector. Cell. 2004;119(1):61–74. [DOI] [PubMed] [Google Scholar]
  • 224. Hashimoto YK, Satoh T, Okamoto M, Takemori H. Importance of autophosphorylation at Ser186 in the A‐loop of salt inducible kinase 1 for its sustained kinase activity. J Cell Biochem. 2008;104(5):1724–1739. [DOI] [PubMed] [Google Scholar]
  • 225. Darling NJ, Cohen P. Nuts and bolts of the salt‐inducible kinases (SIKs). Biochem J. 2021;478(7):1377–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Armouti M, Winston N, Hatano O, Hobeika E, Hirshfeld‐Cytron J, Liebermann J, et al. Salt inducible kinases are critical determinants of female fertility. Endocrinology. 2020;161:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Hers I, Vincent EE, Tavare JM. Akt signalling in health and disease. Cell Signal. 2011;23(10):1515–1527. [DOI] [PubMed] [Google Scholar]
  • 228. Park Y, Maizels ET, Feiger ZJ, Alam H, Peters CA, Woodruff TK, et al. Induction of cyclin D2 in rat granulosa cells requires FSH‐dependent relief from FOXO1 repression coupled with positive signals from Smad. J Biol Chem. 2005;280(10):9135–9148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Liu Z, Rudd MD, Hernandez‐Gonzalez I, Gonzalez‐Robayna I, Fan HY, Zeleznik AJ, et al. FSH and FOXO1 regulate genes in the sterol/steroid and lipid biosynthetic pathways in granulosa cells. Mol Endocrinol. 2009;23(5):649–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Law NC, Weck J, Kyriss B, Nilson JH, Hunzicker‐Dunn M. Lhcgr expression in granulosa cells: roles for PKA‐phosphorylated beta‐catenin, TCF3, and FOXO1. Mol Endocrinol. 2013;27(8):1295–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Dibble CC, Manning BD. Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat Cell Biol. 2013;15(6):555–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Kovacina KS, Park GY, Bae SS, Guzzetta AW, Schaefer E, Birnbaum MJ, et al. Identification of a proline‐rich Akt substrate as a 14‐3‐3 binding partner. J Biol Chem. 2003;278(12):10189–10194. [DOI] [PubMed] [Google Scholar]
  • 233. Wiza C, Nascimento EB, Ouwens DM. Role of PRAS40 in Akt and mTOR signaling in health and disease. Am J Physiol. 2012;302(12):E1453–E1460. [DOI] [PubMed] [Google Scholar]
  • 234. Pallares‐Cartes C, Cakan‐Akdogan G, Teleman AA. Tissue‐specific coupling between insulin/IGF and TORC1 signaling via PRAS40 in drosophila. Dev Cell. 2012;22(1):172–182. [DOI] [PubMed] [Google Scholar]
  • 235. Malla R, Wang Y, Chan WK, Tiwari AK, Faridi JS. Genetic ablation of PRAS40 improves glucose homeostasis via linking the AKT and mTOR pathways. Biochem Pharmacol. 2015;96(1):65–75. [DOI] [PubMed] [Google Scholar]
  • 236. Blancquaert S, Wang LF, Paternot S, Coulonval K, Dumont JE, Harris TE, et al. cAMP‐dependent activation of mammalian target of rapamycin (mTOR) in thyroid cells. Implication in Mitogenesis and activation of CDK4. Mol Endocrinol. 2010;24(7):1453–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Li RH, Ng EH. Management of anovulatory infertility. Best Pract Res Clin Obstet Gynaecol. 2012;26(6):757–768. [DOI] [PubMed] [Google Scholar]
  • 238. Ubaldi F, Vaiarelli A, D'Anna R, Rienzi L. Management of poor responders in IVF: is there anything new? Biomed Res Int. 2014;2014:352098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Sunderam S, Kissin DM, Crawford SB, Folger SG, Jamieson DJ, Warner L, et al. Assisted reproductive technology surveillance—United States, 2014. Morb Mortal Wkly Rep Surveill Summ. 2017;66(6):1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Greenseid K, Jindal S, Hurwitz J, Santoro N, Pal L. Differential granulosa cell gene expression in young women with diminished ovarian reserve. Reprod Sci. 2011;18(9):892–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Bahceci M, Ulug U, Turan E, Akman MA. Comparisons of follicular levels of sex steroids, gonadotropins and insulin like growth factor‐1 (IGF‐1) and epidermal growth factor (EGF) in poor responder and normoresponder patients undergoing ovarian stimulation with GnRH antagonist. Eur J Obstet Gynecol Reprod Biol. 2007;130(1):93–98. [DOI] [PubMed] [Google Scholar]
  • 242. Oosterhuis GJ, Vermes I, Lambalk CB, Michgelsen HW, Schoemaker J. Insulin‐like growth factor (IGF)‐I and IGF binding protein‐3 concentrations in fluid from human stimulated follicles. Hum Reprod. 1998;13(2):285–289. [DOI] [PubMed] [Google Scholar]
  • 243. Gleicher N, Darmon SK, Molinari E, Patrizio P, Barad DH. Importance of IGF‐I levels in IVF: potential relevance for growth hormone (GH) supplementation. J Assist Reprod Genet. 2022;39(2):409–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Goodarzi MO, Jones MR, Chen YD, Azziz R. First evidence of genetic association between AKT2 and polycystic ovary syndrome. Diabetes Care. 2008;31(12):2284–2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Stubbs SA, Webber LJ, Stark J, Rice S, Margara R, Lavery S, et al. Role of insulin‐like growth factors in initiation of follicle growth in normal and polycystic human ovaries. J Clin Endocrinol Metab. 2013;98(8):3298–3305. [DOI] [PubMed] [Google Scholar]

Articles from Reproductive Medicine and Biology are provided here courtesy of John Wiley & Sons Australia, Ltd on behalf of Japan Society for Reproductive Medicine.

RESOURCES