You are on page 1of 3

N EW S A ND V IE W S

Female Fertility: It Takes Two to Tango

Lucy X. Chen1 and Patricia T. Jimenez1


1
Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology, University of Texas
Southwestern Medical Center, Dallas, Texas 75390

Downloaded from https://academic.oup.com/endo/article/158/7/2074/3920684 by guest on 13 October 2021


he ovary is a complex and well-orchestrated organ mechanisms by which FSHR activation in granulosa cells
with important exocrine and endocrine functions result in successful progression of one follicle into the
in sexual reproduction and maintenance of secondary antral phase, whereas other follicles undergo atresia,
sexual characteristics. The germ cells (oocytes) and so- are unclear.
matic cells (granulosa cells, thecal cells, and stromal cells) Ovarian follicles, particularly the dominant follicle,
of the ovary work in a highly integrated manner to release numerous growth factors, including insulinlike
control oocyte development and to secrete sex steroids growth factors (IGFs) and IGF-binding proteins with
and protein hormones. The most important role of the important paracrine/autocrine actions (2, 3). In mice,
ovary is its cyclical maturation of ovarian follicles fol- insulinlike growth factor 1 receptors (IGF1Rs) are se-
lowed by establishment of a dominant follicle (Graafian lectively expressed in granulosa cells of healthy growing
follicle) and subsequent release of an oocyte. This pro- ovarian follicles in a pattern similar to that of FSHR.
cess, termed folliculogenesis, requires a precise sequence Insulinlike growth factor 1 (IGF1) stimulates follicular
and temporal pattern of gene expression by the theca and steroidogenesis (either alone or in synergy with gonad-
granulosa cells and the oocyte. Folliculogenesis can be otropins). Whereas IGF1 knockout mice have primordial
divided into two phases: the gonadotropin-independent follicles, follicle development stops at the preantral stage,
(preantral) and gonadotropin-dependent (antral) pe- notably because of decreased FSHR expression. Thus, in
riods. The preantral phase is controlled mainly by locally humans and rodents, even in the presence of FSH, fol-
produced growth factors through autocrine/paracrine liculogenesis cannot proceed in the absence of IGF1R
mechanisms (1) and can take up to 300 days. The an- activation. The molecular mechanisms mediating these
tral period, on the other hand, is highly dependent on important effects of IGF1 remain largely unknown.
follicle-stimulating hormone (FSH)/luteinizing hormone It has been hypothesized that the role of IGF is to amplify
(LH), and development of follicles to the preovulatory gonadotropin hormonal actions (4). Molecular studies in
stage can take up to 50 days. In mice and humans, in- mice have shown that although FSH does not affect IGF1
activation of the pulsatile release of FSH through mu- expression, IGF1 augments granulosa cell FSHR expres-
tations in the FSHb subunit or FSH receptor gene sion, suggesting that ovarian IGF1 expression serves to
prevents selection of a dominant follicle resulting in enhance granulosa cell FSH responsiveness (5). Zhou et al.
absence of antral follicles. (5) further demonstrated that FSH actions on granulosa cell
A follicular unit is composed of an oocyte, granulosa differentiation in vitro depend on the presence of IGF1 and
cells, and theca cells. Of these, only granulosa cells ex- active IGF1R and that IGF1R inhibition blocks FSH-
press follicle-stimulating hormone receptors (FSHRs) (2). induced follicle growth in immature rats.
FSHR signaling pathways play a fundamental role in There is abundant evidence that the intracellular path-
growth and differentiation of the dominant follicle. They ways activated by FSHR and the IGF1R cooperate closely to
promote follicular fluid formation, cell proliferation, maintain normal granulosa cell function (Fig. 1). FSHR
estradiol production, and LH receptor expression. The activates the cyclic adenosine monophosphate/protein

ISSN Print 0013-7227 ISSN Online 1945-7170 Abbreviations: AKT, protein kinase B; FSH, follicle-stimulating hormone; FSHR, follicle-
Printed in USA stimulating hormone receptor; IGF, insulinlike growth factor; IGF1, insulinlike growth
Copyright © 2017 Endocrine Society factor 1; IGF1R, insulinlike growth factor 1 receptor; IGF1Rgcko, insulinlike growth factor 1
Received 9 May 2017. Accepted 11 May 2017. receptor transcripts in granulosa cells; LH, luteinizing hormone.

For article see page 2309

2074 https://academic.oup.com/endo Endocrinology, July 2017, 158(7):2074–2076 doi: 10.1210/en.2017-00447


doi: 10.1210/en.2017-00447 https://academic.oup.com/endo 2075

Downloaded from https://academic.oup.com/endo/article/158/7/2074/3920684 by guest on 13 October 2021


Figure 1. Proposed molecular pathways by which granulosa cell IGF1R and FSHR collaborate to generate follicular maturity. AKT phosphorylation
requires both IGF1R and FSHR activation and is essential for granulosa cell differentiation, proliferation, and survival. More importantly, failure of
IGF1R to phosphorylate AKT inhibits advancement of folliculogenesis. cAMP, cyclic adenosine monophosphate; CREB, cAMP response element
binding protein; ERK, extracellular signal-regulated kinase; GC, granulosa cell; mTOR, mechanistic target of rapamycin; P, phosphorylation; PI3K,
phosphoinositide 3-kinase; PIP3, phosphatidylinosiitol (3,4,5)-triphosphate; PKA, protein kinase A.

kinase A pathway, phosphoinositide 3-kinase pathway/ crucial for granulosa cell growth and differentiation.
protein kinase B (AKT), and mitogen-activated protein Granulosa cell IGF1R was crucial for escape from apo-
kinase/extracellular signal-regulated kinase pathways ptosis during transition from primary to secondary fol-
that also mediate IGF1 actions (6, 7). licles. IGF1R, therefore, is an important partner with
In this issue of Endocrinology, Baumgarten et al. (8) FSHR for advanced follicle survival. These findings may
demonstrate an obligatory role of IGF1R in the regulation have important implications for ovarian cancer. In-
of survival, proliferation, and differentiation of granulosa creased IGF expression is often an indication of drug
cells during folliculogenesis in vivo. Prior in vivo studies resistance. Further characterization of FSHR/IGFR mo-
were thwarted by the severe defects and short life span in lecular mechanisms may provide novel ovarian cancer
mouse models with globally deficient IGF1 or IGF1R therapies. In addition, it is possible that these finding may
(9). A clever strategy was used to generate granulosa improve our understanding of female fertility, puberty,
cell-specific IGF1R knockout mice. Animals carrying and menopause, as well as allow safer and more effective
both Esr2Cre and Cyp19Cre were used to obtain ovulation induction in women with polycystic ovarian
maximal recombination of the IGF1R floxed allele, syndrome, diminished ovarian reserve, and primary
thereby generating mice with undetectable IGF1R ovarian sufficiency. The role of puberty, aging, ovarian
transcripts in granulosa cells (IGF1Rgcko). Follicular toxins, and steroid hormone signaling pathways on
development did not progress beyond the preantral IGF1R-FHSR crosstalk is a fertile field for research.
stage in IGF1Rgcko mice.
Previously, it was proposed that the role of IGF1 Acknowledgments
was to augment FSHR expression in granulosa cells of
Address all correspondence and requests for reprints to: Patricia
dominant follicles (5). Here, the authors reveal that
T. Jimenez, MD, Department of Obstetrics and Gynecology,
granulosa cell expression of FSHR was similar in wild-
Division of Reproductive Endocrinology, University of Texas
type and IGF1Rgcko mice. Nonetheless, FSH failed to Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas,
stimulate AKT phosphorylation in IGF1Rgcko. Because Texas 75390. E-mail: ptj6305@gmail.com.
AKT phosphorylation is essential for differentiation This work received support from Grant K12 HD000849,
of granulosa cells, the authors hypothesize that this is awarded to the Reproductive Scientist Development Program
the true mechanism by which IGF1-IGF1R signaling is by the Eunice Kennedy Shriver National Institute of Child
2076 Chen and Jimenez IGFR1 Is Crucial for Folliculogenesis Endocrinology, July 2017, 158(7):2074–2076

Health & Human Development and by the Burroughs Well- 3. Wang HS, Chard T. IGFs and IGF-binding proteins in the regulation
come Fund, as part of the Reproductive Scientist Development of human ovarian and endometrial function. J Endocrinol. 1999;
161(1):1–13.
Program.
4. Adashi EY. The IGF family and folliculogenesis. J Reprod Immunol.
Disclosure Summary: The authors have nothing to disclose. 1998;39(1-2):13–19.
5. Zhou P, Baumgarten SC, Wu Y, Bennett J, Winston N, Hirshfeld-
Cytron J, Stocco C. IGF-I signaling is essential for FSH stimulation of
References AKT and steroidogenic genes in granulosa cells. Mol Endocrinol.
2013;27(3):511–523.
1. Williams CJ, Erickson GF. Morphology and physiology of the ovary. 6. Richards JS, Pangas SA. The ovary: basic biology and clinical im-
In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, plications. J Clin Invest. 2010;120(4):963–972.
Hershman JM, Koch C, Korbonits M, McLachlan R, New M, 7. Beauchamp MC, Yasmeen A, Knafo A, Gotlieb WH. Targeting
Purnell J, Rebar R, Singer F, and Vinik A, eds. Endotext. South insulin and insulin-like growth factor pathways in epithelial ovarian
Dartmouth, MA: MDText.com; 2000. https://www.ncbi.nlm.nih. cancer. J Oncol. 2010;2010:257058.

Downloaded from https://academic.oup.com/endo/article/158/7/2074/3920684 by guest on 13 October 2021


gov/books/NBK278951/. Accessed April 18, 2017. 8. Baumgarten SC, Armouti M, Ko CM, Stocco C. IGF1R expression in
2. Hsu SY, Kubo M, Chun SY, Haluska FG, Housman DE, Hsueh AJ. ovarian granulosa cells is essential for steroidogenesis, follicle survival,
Wilms’ tumor protein WT1 as an ovarian transcription factor: and fertility in female mice. Endocrinology. 2017;158(7):2309–2318.
decreases in expression during follicle development and repression 9. Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying
of inhibin-alpha gene promoter. Mol Endocrinol. 1995;9(10): null mutations of the genes encoding insulin-like growth factor I (Igf-1)
1356–1366. and type 1 IGF receptor (Igf1r). Cell. 1993;75(1):59–72.

You might also like