Skip to main content

The insulin-like growth factor and its players: their functions, significance, and consequences in all aspects of ovarian physiology

Abstract

Background

Insulin-like growth factor (IGF) has unique and well-known functions in female fertility, according to documents reporting improved yield of oocytes, reinforced quality of the embryo, and enhanced live births with simultaneous reduction of miscarriage. However, there is no detailed information on the bio-mechanisms linking such clinical differences.

Main body

IGF and its receptors are expressed in a variety of tissues in the reproductive system such as granulosa cells, oocytes, and theca cells. Hence, the development of female gametes may be directly regulated by IGF, thereby affecting gamete quality and so its competence for implantation. IGF is a central player in changing the fate of cells during survival and proliferation through the modulation of leading signaling pathways, including Jak/STAT, MAP kinase/ERK, and PI3K/Akt, and subsequent impacts on steroidogenesis and cell division.

Conclusion

The current review aims to scrutinize the performance of IGF to regulate the normal ovarian, and its impacts on cell signaling pathways and resulting alterations in steroidogenesis and cell proliferation. The function of IGF and its receptor has been reviewed in female fertility at both molecular and biochemical levels.

Background

Fertility and pregnancy refer to the process of releasing an oocyte, fertilizing it with sperm, implanting an embryo in the uterus, and, eventually, the growth of the embryo until birth. Each month, a number of oocytes begin to grow in fluid-filled sacs called follicles inside the ovaries. Ovulation is the process by which one of the oocytes is expelled from the follicle. Ovulation occurs about 2 weeks before menstruation begins. Following releasing of the oocyte from the ovary, the remaining compartments of the follicle become a cyst called the corpus luteum which begins to secrete a hormone that increases the thickness of the endometrium, thus preparing the uterus for the presence of a fertilized egg [1].

The effective and close bidirectional interactions of the oocyte with its neighboring cells, granulosa, during folliculogenesis are required for the later development of the embryo and pregnancy outcome. One of the pivotal functions of granulosa cells, relevant to oocyte competence, accompanied by cooperation with follicle-stimulating hormone (FSH) and its receptor (FSHR), is the estrogen biosynthesis from androgen precursors via the enzyme aromatase. The outcome of this cooperation has a fundamental role in ovarian functions, follicular development, and folliculogenesis, and female infertility can occur due to gene rearrangements, mutations, or aberrant protein production. Since the ovaries do not function, eggs do not mature and are not liberated from the ovaries [2, 3].

The performance of gonadotropins to regulate the granulosa cell ontogeny is well clarified, and the diverse fate of ovarian transplants under the stimulation of measurable gonadotropins indicates the existence of additional mechanisms of intraovarian modulation. According to the results of studies, intraovarian control is probably performed using topical steroid modulation. However, intraovarian peptides may also have the potential to locally modulate follicular growth. In this regard, there is much evidence of the activity of multiple growth factors. Among the potential ontogeny modulators of granulosa cells, insulin-like growth factors (IGF) seem to be a pivotal actor with unique functions. There is evidence that IGF function, in combination with gonadotropins such as FSH and luteinizing hormone (LH), has a synergistic effect on the promotion and proliferation of granulosa and theca cells [4].

Despite studies in this field, different results are found among the researches. This review summarizes the state of the science regarding new mechanism of dynamic control within the ovary in which IGFs can be a pivotal message and granulosa cell as their place to produce, receive, and act.

Main body

Follicle development from preantral to preovulatory stage

The follicular phase is the first step for menstrual cycle. Mechanisms regulating follicle development and growth are controlled by altering the ligand levels (growth factors and hormones). From an endocrine point of view, the mechanisms of central nervous system (CNS), anterior pituitary, and ovary cascade, regulate the folliculogenesis. In fact, gonadotropin-releasing hormone (GnRH) stimulates gonadotroph cells to produce pulsatile LH and FSH. Finally, these hormones regulate follicle development and growth [5,6,7].

Usually, the ovaries in the human beings express a prevailing follicle during each menstrual cycle, which leads to ovulation. The prevailing follicle in any cycle must completely and timely fulfill all processes of folliculogenesis. During these stages, the selected follicle resists all negative episodes that act to eradicate other follicles [5,6,7].

Oocyte support is the basic function of the follicle. At the time of the oocytes production, the ovarian follicles that surround the oocytes develop and transform from the primordial follicle into preovulatory follicles (folliculogenesis). The follicle contains primary oocytes by preovulatory stage, which is blocked in prophase meiosis 1. At the end of the preovulatory phase, the oocyte insists on meiosis and becomes a secondary oocyte, which is blocked in metaphase 2. This mechanism is influenced by the increase of LH hormone [7].

Within development of primary follicle, the granulosa cell communicates with the oocyte through channels made up of connexin proteins, causing dispersion of metabolites, ions, and other compounds, and has a direct effect on the process of folliculation, ovulation, and fertility [5,6,7]. One or more flattened granulosa cells exist in the primary follicle, which surround the oocyte in one layer, and then deformed to a cubic structure and express FSH receptors, which indicates the beginning of primary follicle development. FSH receptor expression stimuli include FSH, activin, cyclic adenosine monophosphate (cAMP), and TGF. The oocyte genome is also activated, and transcription of genes begins, and the primary paracrine messenger pathways are formed, which are essential for communication between the follicle cells and the oocyte [5,6,7].

The formation of a secondary follicle requires more granulosa cell accumulation, which forms several layers around the oocyte, accompanied by the transition of the stroma-like theca cells to construct two layers of external and internal theca cells. The receptors for hormone LH are expressed by the theca cells. The LH can trigger the synthesis of androgens from theca cells that are converted to estrogen via the granulosa cells and a series of biochemical changes and aromatization. As a result, estrogen levels increase [5,6,7]. Following the elevated FSH level, the antral follicles produce inhibin and estrogen, with negative feedback on FSH. The follicles with smaller number of FSH receptors are unable to grow more and will gradually degenerate. Eventually, only one follicle will survive, and it will be called the dominant follicle. This follicle grows rapidly, reaching 18 to 23 mm in diameter, to become a preovulatory follicle. Finally, after the final follicle is selected, a sudden increase of FSH and LH stimulates the cells of the various follicle components to induce messages that collectively lead to ovulation [7, 8].

An overview on the insulin-like growth factors (IGFs) functions in granulosa cells

There is evidence that IGF function, in combination with gonadotropins such as FSH and LH, has a synergistic effect on the proliferation and promotion of theca and granulosa cells. Based on animal studies, treatment with IGF and FSH increases the differentiation of granulosa cells in rat ovaries, while no alteration is seen in proliferation [9]. In contrast, the augmentation of IGF to rat follicles leads to an increase in both granulosa and theca cells [10]. Nevertheless, it is unclear whether IGF promotes cell proliferation directly via the IGF receptors expressed in them or whether it occurs indirectly by triggering granulosa cell-secreted secondary growth factors, thereby influencing the theca cells directly [10, 11]. IGF-mediated proliferation process of theca cell has been verified in ex vivo model of sheep, to the extent that high IGF levels led to theca cell overgrowth [12]. This impact is validated in vitro, where IGF levels were utilized to assess the damaging effect on rat perinatal follicles, potentially due to stromal cell and theca growth and nutrient depletion [13]. IGF supplementation of the alginate-based growth medium containing bovine secondary follicles resulted in increased E2 synthesis. Furthermore, increased E2 production can maintain follicular structure and performance, leading in greater follicle growth [14] (Fig. 1).

Fig. 1
figure 1

IGF functions in ovarian cells (theca and granulosa)

Interestingly, metabolic disorders and elevated oxidative stress disturb the insulin-associated signaling within the ovary and can dysregulate the dynamics of the ovarian function through the impair the ovarian reserve, survival, and quality of the eggs [15]. In women with PCOS, insulin resistance with compensatory hyperinsulinemia induces androgen overproduction in the ovaries, which can lead to hyperandrogenism. Insulin acts on ovarian androgen production via IGF type 1 receptors [16].

Expression of IGF in primates and rodents

Based on previous findings, the FSH as an essential agent has a role to generate the follicles (before ovulation) and differentiate the granulosa cells, but the presence of FSH alone is not enough, and other factors are necessary for growth and differentiation. Some studies have shown that in rodents that lack FSH or FSH receptors, follicular progression is blocked beyond the preantral stage [3, 4]. A block exists on the production of preovulatory follicle in mice with IGF-1 deficiency [5], highlighting a need for active IGF system in the maturation of follicle until the phase of pre-ovulation. The stimulation of CYP19A1 secretion via FSH requires the activity of IGF-1 receptor (IGF-1R) in humans and rodents [6, 7]. Hence, the follicular transition from preantral stage to preovulatory phase needs the IGF and FSH signaling pathways. However, detailed information is not available on how these processes are regulated by FSH and IGFs, especially in humans.

Humans and rodents have significant differences in the IGF system of their ovaries. Granulosa cells in the rodents are more responsible for the synthesis of IGF-1 [8]. Conversely, the granulosa cells in human have higher levels of IGF-2 mRNA, but not detectable IGF-1 mRNA [7, 9, 10]. As a result, the follicular fluid of prevailing follicles in humans contains IGF-2 up to 10 times higher than IGF-1 [11, 12]. Greater intrafollicular IGF-2 levels can explain why a 1.6-fold elevation in intrafollicular IGF-1 lacks any impact on follicular maturation in primates [13]. However in the 3-D culture medium, adding IGF-1 can enhance the production of mature oocytes, and injection of IGF-1 before ovulation induction will increase the count of retrieved oocytes [17].

The positive correlation of intrafollicular IGF-2 levels with oocyte maturation and follicular maturation confirms the essential function of IGF-2 in the regulation of follicular growth in humans [12, 14, 15]. Based on this evidence, the IGF-2 has a pivotal function in the IGF system in the granulosa cells in humans. Previous observations also highlighted the stimulatory role of IGF-2 in the proliferation [18] and expression of progesterone and estradiol in luteinized granulosa cells in humans [19, 20]. The IGF-2 in primates can enhance the steroidogenesis and accelerate the synthesis of vascular endothelial growth factor in the preovulatory follicles [19, 21, 22]. The aggregation of progesterone and estradiol is triggered by the IGF-2 in granulosa cells of small (2–7 mm) and preovulatory follicles, while the IGF-2 does not impact on the proliferation [22]. The axial function of IGF-2 in follicular maturation through autocrine actions is confirmed via preferential gene expression and IGF-2 production by prevailing follicle granulosa cells in humans [9].

Interaction of IGF with FSH and LH

Simultaneous effect of LH and FSH on their gonadotropin supplementation receptors reduces signaling, which is essential for differentiation, proliferation, and steroidogenesis, and the two signals increase E2 and P4 production through cAMP and PKA. The two LHR and FSHR are receptors associated with G protein, which transmit intracellular cascade through cAMP adenylate cyclase activity, and PKA activation, after which the CREB transcription factor is phosphorylated. cAMP response element-binding protein (CREB) transcribes various genes by binding to cAMP in DNA (including aromatase and sex steroids precursor). In the granulosa cells, the interaction of GH with GHR modulates the function of FSH, as well as induces LHR [9, 23]. Such LHR generation is the major indicator for the differentiation of granulosa cells and theca cells, can be affected by the ovarian induction of IGF, while proliferating granulosa cells, and acts as a paracrine. IGF can alter and enhance the sensitivity of theca or granulosa cells to stimulation of gonadotropin, and thus regulate the secretion of sex steroid, and liberation into follicles, finally leading to cell growth enhancement s paracrine/autocrine steroidization factors. According to two-cell theory, steroidogenesis is regulated in the ovary based on dependent and independent procedures, so that the theca cells are triggered by LH to generate androgens that are converted to diverse estrogens due to the expressed aromatase in the granulosa cells [14]. Interestingly, investigations on females with diminished ovarian reserve have shown that the supplementation with IGF increases LHR, FSHR, and GHR expression in isolated human granulosa cells. IGF supports the maturation process of luteinization because of increasing LHR density and decreasing preovulatory FSHR expression. Cytosolic aggregation of Camp and PKA signaling activation can be initiated by the simultaneous effect of GHR and IGFR, which possibly affects the gonadotropin response [18, 19].

IGF-1, like GH, triggers the theca and granulosa cell differentiation and proliferation via the enhancement of the FSH function in granulosa cells. It has been shown that IGFR is absolutely necessary to induce PI3K/Akt pathway and differentiate granulosa cells by FSH [20, 21, 24, 25]. In a study by Zhao et al., the primary follicles were triggered by IGF-1 through the measurement of DNA increase in follicular cells. In their study, IGF-1-cultured cells have preferred morphology because of elevated count of junctions between the granulosa cells and theca cells as well as the granulosa cells and oocytes. Moreover, in this study, the availability of IGF-1 and FSH enhanced the growth of preantral follicles following the FSHR activation [26]. Animal studies using gene knockouts have implied a more direct and greater impact of IGF than GH on reproduction [22]. In this context, female rats with completely sterile IGF-1R knockout had no antral follicles and exhibited a decrease up to 90% in the serum E2 level [27]. Inactivating IGF-R or IGF-1 by knockout is not mostly compatible with life. The knockout rat by IGF-1 significantly reduced FSH receptor expression, thereby reducing aromatase expression and E2 secretion, which leads to infertility in both genders in some cases. Under in vitro condition, Magalhaes-Padiha et al. showed greater growth of triggered preantral follicles by IGF-1, probably because of cell proliferation, since IGF-1 can enhance the granulosa cell nuclear maturation in preantral follicle [22].

The expression of gonadotropin receptor might be regulated by IGF signaling. IGFR and FSH in combination can result in different intracellular signaling pathways like cAMP formation, thereby triggering CREB and PKA alongside induction of PI3K/Akt and MAPK/ERK1/2 pathways. Such signaling mechanisms elevate the activity of aromatase and production of LHR. IGF-1 or 2 and FSH act together and interact with corresponding receptors to create aromatase activity. IGF-1 exerts unique and stimulatory influences on the granulosa cells and according to reports increases the production of steroidogenic CYP19A1, 3-beta-hydroxysteroid dehydrogenase, CYP11A1, and the expression of IGF-1R and FSHR. Considering to results, IGF-1 activates steroidogenic regulatory genes and apoptosis by activating the PI3K/Akt signaling in the bovine granulosa cell. IGF-1/2 can both trigger secretion of sex steroids implicated in the growth of follicles [14, 28,29,30,31].

The combination of IGF-1 and LH enhances P granulosa cells and has a regulatory impact on the production of E2 in luteal cells. Significantly, IGF-1R plays an important role to increase FSH-stimulated StAR expression, as an essential factor for cholesterol transport to mitochondria, which is the first stair to produce sex steroids and pregnenolone. In addition, according to reports, great IGF level impedes the performance of anti-Mullerian hormone (AMHAMH) that is exclusively secreted in the gonadal tissues. AMH is transforming growth factor beta (TGF-B) belonging to the family growth hormone, which decreases growth and performance of antral and preantral follicles in mammals. This issue may clear the performance of IGF to regulate follicle selection and growth. Based on these results, the IGF is a prominent actor to regulate the growth of follicle via the proliferation and differentiation of granulosa cells, production of steroid, and gonadotropin-stimulating activity. Table 1 summarizes these associated effects [14, 31,32,33].

Table 1 The involvement of IGF in ovarian physiology and infertility

Expression, regulation, and secretion of IGF

The GHR-GH interaction activates conventional and unconventional signaling pathways. In conventional one, the pituitary gland triggers hepatic GH cells to liberate GHR into circulation via transcription factors induced by IGF, GH, and GHR. The interactions of ligand receptor lead to adsorption and autophosphorylation in the GHR cytoplasmic domain, and the JAK2/GHR complex subsequently phosphorylates STAT molecules, modifies gene transcription, and significantly results in influences on the cell proliferation. STAT5b can more significantly and directly regulate the production of IGF-1 and also mediate the production of GH-induced IGF-1 in the granulosa cells of rats. Unconventional signaling usually has no dependence on JAK2 and includes using non-receptor tyrosine kinases, CY phospholipase stimulation, and organelle cytosolic calcium flow.

It is unclear which system or hormone has significance since the ovarian performance is affected by systemic IGF and GH, GH-independent IGF, or GH-induced peripheral IGF. Nevertheless, the GH-IGF axis as a pivotal growth factor is implicated in folliculogenesis. The IGF-1 mRNA expression is increased by the GH in the precursor follicles of rats, leading to the IGF-1 synthesis from sheep granulosa cell. In addition, IGFBP-3 counteracts anti-apoptotic influence of GH, underlining the elevation of locally IGF-1 synthesis by the exogenous GH and subsequently resulting in increased survival of follicle. As a result, the ovarian GH-IGF interaction is complex because it is used by granulosa cells in autocrine and paracrine procedures and needs no stimulation of GH [14, 39,40,41,42,43].

Systemic IGF contains IGF-1 and -2, corresponding receptors, and six IGFBPs, which regulate the bioavailability of IGF. The paracrine synthesis of such members is of great significance, while sheep folliculogenesis, and peripheral IGF, elevates due to the decrease of binding proteins expression. While IGF-2R and IGFBP-5 increase, IGF-2 expression decreases in atretic follicles. Based on these findings, reduced peripheral bioavailability of IGF leads to follicle elimination, and the synthesis of IGF is pivotal for the survival of follicles. Ovarian IGF levels are also related to the folliculogenesis stage. Hence, there is low level of IGF in theca cells resulting from follicles medium in size, also in neonatal ova, while IGF-2 expression is high in the isolated granulosa cells from antral follicles. Thus, apparently, a dynamic performance can be seen for the activity of IGF and GH because of follicle maturation and growth [37, 44,45,46,47,48,49,50,51].

IGF signaling pathway in granulosa cells during human ovarian follicle development

Despite numerous differences in the reproductive systems of men and women, the behavior versus gonadotropins, such as the synthesis and secretion of sex steroids and cell proliferation, is highly similar in both systems. According to studies, it is possible that any effect of IGF applies its effect by affecting the synthesis pathways, secretion of sex steroids and cell proliferation, and enhancing steroidogenesis, followed by the synthesis of products that exert their specific influences on the cell survival and proliferation [14].

The cAMP/PKA pathway is triggered by the FSH, because of stimulating FSHR, and subsequently, CREB mediates the transcription of diverse genes. Such a trend increases the production of proteins related to steroidogenesis like StAR, aromatase, and LHR [52, 53]. CREB directly regulates the aromatase gene [54], which produces estrogens by converting androgens, whereas the StAR causes the mitochondrial transport of cholesterol for the synthesis of testosterone, P4 and E2, in steroid cells. Besides these events, activation of FSHR and LHR GPCR can lead to other main cell signaling events, which affect steroidogenesis. One of these pathways is pivotal route of PI3K/Akt cascade, as is an obvious regulatory mechanism for cell survival, proliferation, and metabolism [55], which is directly stimulated via FSHR, after directly interacting with adapter proteins of 14-3-3Ï„ [56]. LH can also stimulate the PI3K/Akt pathway, and its activity is increased due to FSH [57, 58].

The multifunctional signaling center of Akt regulates cell proliferation, metabolism, and mortality [14, 59]. Akt is activated by FSH, which is needed to produce α-inhibin, CYP19, 3β-HSD, and LHR [60]. Evidence suggests the production of FSHR-induced aromatase needed for activation of PI3K/Akt and cAMP/PKA mechanisms [14, 61]. Recent studies on granulosa cells in rodent and human confirmed the intact IGF-1R signaling needed for FSHR-induced Akt phosphorylation [27, 60]. It also appears that the FSHR function necessarily requires PI3K/Akt signaling and is supported by the stimulation of IGF-IGFR Akt. Based on multiple investigations, the FSH is unable to increase the expression of LHR, CYP19, and STAR in exposure to the IGF inhibitors [60]. Intracellular IGF signaling connects to FSHR and LHR signaling by setting PI3K/Akt cascade. Proinsulin and IGF have almost the same structure, with both IGFR and insulin (IR) receptors binding. The IGF-IR interaction results in phosphorylation and adsorption of insulin receptor 1 (IRS1) or insulin receptor 2 (IRS2) by activating PI3K and then Akt [14].

One of the advantages of triggered IGF in PLC/PKC pathway is the enhancement of CREB-induced transcription. IGFR can directly activate Cγ (PLCγ), followed by phospholipid hydrolysis to produce diacylglycerol, or DAG, and inositol-4,4,4-triphosphate, or IP3 [43]. IP3 increases the cytosolic calcium flux of the organelles, and DAG also activates PKC. PKC can directly stimulate CREB-mediated gene transcription such as StAR and/or aromatase [62]. According to studies, PKC stimulation increases the expression of StAR and leads to production of progesterone in granulosa cells [62, 63]. It is noteworthy that FSHR can activate the PKC pathway, which occurs through the formation of IP3 and DAG by FSHR, followed by the cumulus cell proliferation and the oocyte meiotic maturation [64], which indicates the convergence between IGF signaling and gonadotropin GPCRs. The connection between these signaling systems in part justifies ex vivo or in vitro physiological influences.

When MAPK/ERK1/2 and p38 MAPK signaling pathways begin, the result is the common denominator of signaling pathways, which alter gene expression, metabolism, and cell proliferation [43]. The function of ERK1/2 leads to an increase in cellular mitogenic messages, which is indirectly triggered by PKA through the increased intracellular calcium induced by PLC/PKC events, both of which, according to the description, have been stimulated by IGF activity [53, 65]. The p38 MAPK in the granulosa cells is implicated in production of pro-apoptotic signals [65]. Nevertheless, there is limited information about the behavior of MAPK/ERK1/2 pathway in steroidogenesis [53, 66]. However, in some studies, stimulation of IGF-mediated progesterone production in the ovarian cells in human depends on p38 MAPK and MAPK/ERK1/2 pathways [67]. The PI3K/Akt and pro-MAPK/ERK1/2 signaling is triggered by IGF, and the activation of IGF-1R occurs in mass granulosa cells [21] (Fig. 2).

Fig. 2
figure 2

IGF signaling in theca and granulosa cells. Created with BioRender.com

Conclusion

Ovarian follicles, along with the oocyte and supporting cells, determine the female reproductive cycle and thus fertility. Endogenous variables, auto-, para-, and endocrine mechanisms, all impact ovarian function. The main function of granulosa cells, relevant to oocyte development and ovarian functions, is steroidogenesis, for which different signaling pathways, including Jak/STAT, PI3K/Akt, and pro-MAPK/ERK1/2, regulate this fundamental process, all of which have a shared modulator named IGF. IGF and its receptor are found in reproductive tissues and cells and may directly govern gamete development and influence its quality.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

FSH:

Follicle-stimulating hormone

CNS:

Central nervous system

GnRH:

Gonadotropin-releasing hormone

cAMP:

Cyclic adenosine monophosphate

IGF:

Insulin-like growth factors

CREB:

cAMP response element-binding protein

hCG:

Human chorionic gonadotropin

PI3K/Akt:

Phosphatidylinositol 3-kinase/protein kinase B

MAPK/ERK1:

Mitogen-activated protein kinase/extracellular signal-regulated kinases

CYP19A1:

Cytochrome P450 family 19 subfamily A member 1

References

  1. Adamson GD, Rutherford AJ (2018) Commercialization of in-vitro fertilization. In: In-Vitro Fertilization: The Pioneers’ History

    Google Scholar 

  2. Henríquez S, Kohen P, Muñoz A, Godoy A, Orge F, Strauss JF III et al (2017) In-vitro study of gonadotrophin signaling pathways in human granulosa cells in relation to progesterone receptor expression. Reprod Biomed Online 35(4):363–371

    Article  PubMed  CAS  Google Scholar 

  3. Willis D, Mason H, Gilling-Smith C, Franks S (1996) Modulation by insulin of follicle-stimulating hormone and luteinizing hormone actions in human granulosa cells of normal and polycystic ovaries. J Clin Endocrinol Metabol 81(1):302–309

    CAS  Google Scholar 

  4. Adashi EY, Resnick CE, D'Ercole AJ, Svoboda ME, van Wyk JJ (1985) Insulin-like growth factors as intraovarian regulators of granulosa cell growth and function. Endocr Rev 6(3):400–420

    Article  CAS  PubMed  Google Scholar 

  5. Macklon NS, Fauser BC (2001) Follicle-stimulating hormone and advanced follicle development in the human. Arch Med Res 32(6):595–600

    Article  CAS  PubMed  Google Scholar 

  6. Johnson A, Woods DC (2007) Ovarian dynamics and follicle development. Reprod Biol Phylogeny Birds 6:243–277

    Google Scholar 

  7. Baerwald AR, Adams GP, Pierson RA (2012) Ovarian antral folliculogenesis during the human menstrual cycle: a review. Hum Reprod Update 18(1):73–91

    Article  PubMed  Google Scholar 

  8. Erickson GF, Shimasaki S (2000) The role of the oocyte in folliculogenesis. Trends Endocrinol Metab 11(5):193–198

    Article  CAS  PubMed  Google Scholar 

  9. Jia X-C, Kalmijn J, Hsueh AJ (1986) Growth hormone enhances follicle-stimulating hormone-induced differentiation of cultured rat granulosa cells. Endocrinology. 118(4):1401–1409

    Article  CAS  PubMed  Google Scholar 

  10. Kobayashi J, Mizunuma H, Kikuchi N, Liu X, Andoh K, Abe Y et al (2000) Morphological assessment of the effect of growth hormone on preantral follicles from 11-day-old mice in an in vitro culture system. Biochem Biophys Res Commun 268(1):36–41

    Article  CAS  PubMed  Google Scholar 

  11. Sirotkin A, Makarevich A (1999) GH regulates secretory activity and apoptosis in cultured bovine granulosa cells through the activation of the cAMP/protein kinase A system. J Endocrinol 163(2):317–328

    Article  CAS  PubMed  Google Scholar 

  12. Arunakumari G, Shanmugasundaram N, Rao V (2010) Development of morulae from the oocytes of cultured sheep preantral follicles. Theriogenology. 74(5):884–894

    Article  CAS  PubMed  Google Scholar 

  13. Zhao J, Van Tol H, Taverne M, Van der Weijden G, Bevers M, Van den Hurk R (2000) The effect of growth hormone on rat pre-antral follicles in vitro. Zygote. 8(3):275–283

    Article  CAS  PubMed  Google Scholar 

  14. Ipsa E, Cruzat VF, Kagize JN, Yovich JL, Keane KN (2019) Growth hormone and insulin-like growth factor action in reproductive tissues. Front Endocrinol 10:777

    Article  Google Scholar 

  15. Dri M, Klinger FG, De Felici M (2021) The ovarian reserve as target of insulin/IGF and ROS in metabolic disorder-dependent ovarian dysfunctions. Reprod Fertil 2(3):R103–Rr12

    Article  PubMed  PubMed Central  Google Scholar 

  16. Firmansyah A, Chalid MT, Farid RB, Nusratuddin N (2018) The correlation between insulin-like growth factor binding protein 1 (IGFBP-1) and homeostasis model assessment of insulin resistance (HOMA-IR) in polycystic ovarian syndrome with insulin resistance. Int J Reprod Biomed 16(11):679–682

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Dai S, Zhang H, Yang F, Shang W, Zeng S (2022) Effects of IGF-1 on the three-dimensional culture of ovarian preantral follicles and superovulation rates in mice. Biology (Basel) 11(6)

  18. Regan SL, Knight PG, Yovich JL, Arfuso F, Dharmarajan A (2018) Growth hormone during in vitro fertilization in older women modulates the density of receptors in granulosa cells, with improved pregnancy outcomes. Fertil Steril 110(7):1298–1310

    Article  CAS  PubMed  Google Scholar 

  19. Bergan-Roller HE, Sheridan MA (2018) The growth hormone signaling system: insights into coordinating the anabolic and catabolic actions of growth hormone. Gen Comp Endocrinol 258:119–133

    Article  CAS  PubMed  Google Scholar 

  20. Zeleznik AJ, Saxena D, Little-Ihrig L (2003) Protein kinase B is obligatory for follicle-stimulating hormone-induced granulosa cell differentiation. Endocrinology. 144(9):3985–3994

    Article  CAS  PubMed  Google Scholar 

  21. Baumgarten SC, Convissar SM, Fierro MA, Winston NJ, Scoccia B, Stocco C (2014) IGF1R signaling is necessary for FSH-induced activation of AKT and differentiation of human Cumulus granulosa cells. J Clin Endocrinol Metabol 99(8):2995–3004

    Article  CAS  Google Scholar 

  22. Magalhães-Padilha D, Duarte A, Araújo V, Saraiva M, Almeida A, Rodrigues G et al (2012) Steady-state level of insulin-like growth factor-I (IGF-I) receptor mRNA and the effect of IGF-I on the in vitro culture of caprine preantral follicles. Theriogenology. 77(1):206–213

    Article  PubMed  CAS  Google Scholar 

  23. Nimrod A (1981) The induction of ovarian LH-receptors by FSH is mediated by cyclic AMP. FEBS Lett 131(1):31–33

    Article  CAS  PubMed  Google Scholar 

  24. Schams D, Berisha B, Kosmann M, Amselgruber W (2002) 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 22(1):51–72

    Article  CAS  PubMed  Google Scholar 

  25. BK C, Scaramuzzi R, Webb R (1995) Control of antral follicle development and selection in sheep and cattle. J Reprod Fertil Suppl 49:335–350

    Google Scholar 

  26. Zhao J, Taverne M, Van Der Weijden G, Bevers M, Van Den Hurk R (2001) Insulin-like growth factor-I (IGF-I) stimulates the development of cultured rat pre-antral follicles. Mol Reprod Dev: Incorp Gamete Res 58(3):287–296

    Article  CAS  Google Scholar 

  27. Baumgarten SC, Armouti M, Ko C, Stocco C (2017) IGF1R expression in ovarian granulosa cells is essential for steroidogenesis, follicle survival, and fertility in female mice. Endocrinology. 158(7):2309–2318

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sun GW, Kobayashi H, Suzuki M, Kanayama N, Terao T (2003) 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. 144(3):793–801

    Article  CAS  PubMed  Google Scholar 

  29. Mani AM, Fenwick MA, Cheng Z, Sharma MK, Singh D, Wathes DC (2010) IGF1 induces up-regulation of steroidogenic and apoptotic regulatory genes via activation of phosphatidylinositol-dependent kinase/AKT in bovine granulosa cells. Reproduction. 139(1):139

    Article  CAS  PubMed  Google Scholar 

  30. Kranc W, Budna J, Kahan R, Chachuła A, Bryja A, Ciesiółka S et al (2017) Molecular basis of growth, proliferation, and differentiation of mammalian follicular granulosa cells. J Biol Regul Homeost Agents 31(1):1–8

    CAS  PubMed  Google Scholar 

  31. Andreassen M, Frystyk J, Faber J, Kristensen L, Juul A (2013) Growth hormone (GH) activity is associated with increased serum oestradiol and reduced anti-M üllerian hormone in healthy male volunteers treated with GH and a GH antagonist. Andrology. 1(4):595–601

    Article  CAS  PubMed  Google Scholar 

  32. Xu J, Bishop C, Lawson M, Park B, Xu F (2016) Anti-Müllerian hormone promotes pre-antral follicle growth, but inhibits antral follicle maturation and dominant follicle selection in primates. Hum Reprod 31(7):1522–1530

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yang M, Cushman R, Fortune J (2017) Anti-Müllerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles. MHR: Basic Sci Reprod Med 23(5):282–291

    CAS  Google Scholar 

  34. Waters M, Hoang H, Fairlie D, Pelekanos R, Brown R (2006) New insights into growth hormone action. J Mol Endocrinol 36(1):1–7

    Article  CAS  PubMed  Google Scholar 

  35. Zhou H, Zhang Y (2005) Effect of growth factors on in vitro development of caprine preantral follicle oocytes. Anim Reprod Sci 90(3-4):265–272

    Article  CAS  PubMed  Google Scholar 

  36. Zhou J, Kumar TR, Matzuk MM, Bondy C (1997) Insulin-like growth factor I regulates gonadotropin responsiveness in the murine ovary. Mol Endocrinol 11(13):1924–1933

    Article  CAS  PubMed  Google Scholar 

  37. Hastie PM, Haresign W (2006) Expression of mRNAs encoding insulin-like growth factor (IGF) ligands, IGF receptors and IGF binding proteins during follicular growth and atresia in the ovine ovary throughout the oestrous cycle. Anim Reprod Sci 92(3-4):284–299

    Article  CAS  PubMed  Google Scholar 

  38. Guthrie H, Garrett W, Cooper B (1998) Follicle-stimulating hormone and insulin-like growth factor-I attenuate apoptosis in cultured porcine granulosa cells. Biol Reprod 58(2):390–396

    Article  CAS  PubMed  Google Scholar 

  39. Rotwein P (2012) Mapping the growth hormone–Stat5b–IGF-I transcriptional circuit. Trends Endocrinol Metab 23(4):186–193

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Takahashi Y (2017) The role of growth hormone and insulin-like growth factor-I in the liver. Int J Mol Sci 18(7):1447

    Article  PubMed Central  CAS  Google Scholar 

  41. Dehkhoda F, Lee CM, Medina J, Brooks AJ (2018) The growth hormone receptor: mechanism of receptor activation, cell signaling, and physiological aspects. Front Endocrinol 9:35

    Article  Google Scholar 

  42. Nakamura E, Otsuka F, Inagaki K, Miyoshi T, Matsumoto Y, Ogura K et al (2012) Mutual regulation of growth hormone and bone morphogenetic protein system in steroidogenesis by rat granulosa cells. Endocrinology. 153(1):469–480

    Article  CAS  PubMed  Google Scholar 

  43. Rowlinson SW, Yoshizato H, Barclay JL, Brooks AJ, Behncken SN, Kerr LM et al (2008) An agonist-induced conformational change in the growth hormone receptor determines the choice of signalling pathway. Nat Cell Biol 10(6):740–747

    Article  CAS  PubMed  Google Scholar 

  44. Devesa J, Caicedo D (2019) The role of growth hormone on ovarian functioning and ovarian angiogenesis. Front Endocrinol 10:450

    Article  Google Scholar 

  45. Matsuda F, Inoue N, Manabe N, Ohkura S (2012) Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J Reprod Dev 58(1):44–50

    Article  CAS  PubMed  Google Scholar 

  46. Eisenhauer KM, Chun S-Y, Billig H, Hsueh AJ (1995) Growth hormone suppression of apoptosis in preovulatory rat follicles and partial neutralization by insulin-like growth factor binding protein. Biol Reprod 53(1):13–20

    Article  CAS  PubMed  Google Scholar 

  47. Khalid M, Haresign W, Luck M (2000) Secretion of IGF-1 by ovine granulosa cells: effects of growth hormone and follicle stimulating hormone. Anim Reprod Sci 58(3-4):261–272

    Article  CAS  PubMed  Google Scholar 

  48. Humbel RE (1990) Insulin-like growth factors I and II. Eur J Biochem 190(3):445–462

    Article  CAS  PubMed  Google Scholar 

  49. Rajaram S, Baylink DJ, Mohan S (1997) Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev 18(6):801–831

    CAS  PubMed  Google Scholar 

  50. Monget P, Bondy C (2000) Importance of the IGF system in early folliculogenesis. Mol Cell Endocrinol 163(1-2):89–93

    Article  CAS  PubMed  Google Scholar 

  51. Zho J, Bondy C (1993) Anatomy of the human ovarian insulin-like growth factor system. Biol Reprod 48(3):467–482

    Article  Google Scholar 

  52. Sarit F, Ada D (2005) Ovarian transcriptomes as a tool for a global approach of genes modulated by gonadotropic hormones in human ovarian granulosa cells. Endocrine. 26(3):259–265

    Article  CAS  PubMed  Google Scholar 

  53. Casarini L, Crépieux P (2019) Molecular mechanisms of action of FSH. Front Endocrinol 10:305

    Article  Google Scholar 

  54. Carlone DL, Richards JS (1997) 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 11(3):292–304

    CAS  PubMed  Google Scholar 

  55. Chen Y-J, Hsiao P-W, Lee M-T, Mason JI, Ke F-C, Hwang J-J (2007) Interplay of PI3K and cAMP/PKA signaling, and rapamycin-hypersensitivity in TGFβ1 enhancement of FSH-stimulated steroidogenesis in rat ovarian granulosa cells. J Endocrinol 192(2):405–419

    Article  CAS  PubMed  Google Scholar 

  56. Cohen BD, Nechamen CA, Dias JA (2004) Human follitropin receptor (FSHR) interacts with the adapter protein 14-3-3τ. Mol Cell Endocrinol 220(1-2):1–7

    Article  CAS  PubMed  Google Scholar 

  57. Casarini L, Santi D, Brigante G, Simoni M (2018) Two hormones for one receptor: evolution, biochemistry, actions, and pathophysiology of LH and hCG. Endocr Rev 39(5):549–592

    Article  PubMed  Google Scholar 

  58. Casarini L, Lispi M, Longobardi S, Milosa F, La Marca A, Tagliasacchi D et al (2012) LH and hCG action on the same receptor results in quantitatively and qualitatively different intracellular signalling. PLoS One 7(10):e46682

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hsueh AJ, Adashi E, Jones PB, Welsh TH Jr (1984) Hormonal regulation of the differentiation of cultured ovarian granulosa cells. Endocr Rev 5(1):76–127

    Article  CAS  PubMed  Google Scholar 

  60. Zhou P, Baumgarten SC, Wu Y, Bennett J, Winston N, Hirshfeld-Cytron J et al (2013) IGF-I signaling is essential for FSH stimulation of AKT and steroidogenic genes in granulosa cells. Mol Endocrinol 27(3):511–523

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Donadeu FX, Ascoli M (2005) The differential effects of the gonadotropin receptors on aromatase expression in primary cultures of immature rat granulosa cells are highly dependent on the density of receptors expressed and the activation of the inositol phosphate cascade. Endocrinology. 146(9):3907–3916

    Article  CAS  PubMed  Google Scholar 

  62. Manna PR, Huhtaniemi IT, Stocco DM (2009) Mechanisms of protein kinase C signaling in the modulation of 3′, 5′-cyclic adenosine monophosphate-mediated steroidogenesis in mouse gonadal cells. Endocrinology. 150(7):3308–3317

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Manna PR, Soh J-W, Stocco DM (2011) The involvement of specific PKC isoenzymes in phorbol ester-mediated regulation of steroidogenic acute regulatory protein expression and steroid synthesis in mouse Leydig cells. Endocrinology. 152(1):313–325

    Article  CAS  PubMed  Google Scholar 

  64. Yamashita Y, Okamoto M, Ikeda M, Okamoto A, Sakai M, Gunji Y et al (2014) Protein kinase C (PKC) increases TACE/ADAM17 enzyme activity in porcine ovarian somatic cells, which is essential for granulosa cell luteinization and oocyte maturation. Endocrinology. 155(3):1080–1090

    Article  PubMed  CAS  Google Scholar 

  65. Shiota M, Sugai N, Tamura M, Yamaguchi R, Fukushima N, Miyano T et al (2003) Correlation of mitogen-activated protein kinase activities with cell survival and apoptosis in porcine granulosa cells. Zoolog Sci 20(2):193–201

    Article  CAS  PubMed  Google Scholar 

  66. Moore RK, Otsuka F, Shimasaki S (2001) Role of ERK1/2 in the differential synthesis of progesterone and estradiol by granulosa cells. Biochem Biophys Res Commun 289(4):796–800

    Article  CAS  PubMed  Google Scholar 

  67. Seto-Young D, Avtanski D, Varadinova M, Park A, Suwandhi P, Leiser A et al (2011) Differential roles of MAPK-Erk1/2 and MAPK-p38 in insulin or insulin-like growth factor-I (IGF-I) signaling pathways for progesterone production in human ovarian cells. Horm Metab Res 43(06):386–390

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

PA and AA contributed to the design of the study, interpretation of data, and drafted the manuscript. EH was involved in editing the manuscript and approved the final submission. MB was involved in revising the manuscript and approved the final submission. All authors reviewed the final manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Mahshid Bazrafkan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Afradiasbagharani, P., Hosseini, E., Allahveisi, A. et al. The insulin-like growth factor and its players: their functions, significance, and consequences in all aspects of ovarian physiology. Middle East Fertil Soc J 27, 27 (2022). https://doi.org/10.1186/s43043-022-00119-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43043-022-00119-1

Keywords