Elsevier

Gene

Volume 753, 30 August 2020, 144812
Gene

Review
Molecular insights into hormone regulation via signaling pathways in Sertoli cells: With discussion on infertility and testicular tumor

https://doi.org/10.1016/j.gene.2020.144812Get rights and content

Highlights

  • Sertoli cells act as a protector, nourisher, and supporter for spermatogenesis.

  • The hypothalamic-pituitary–gonadal axis is related with pathways in Sertoli cells.

  • The AR, cAMP/PKA and PI3k/Akt pathways in Sertoli cells mediate spermatogenesis.

  • The pathways are abnormal in infertile or testicular cancer patients.

Abstract

Spermatogenesis is a complex and elaborate differentiation process and is critical for male fertility. The hypothalamic-pituitary–gonadal axis serves as a significant neuroendocrine system to regulate spermatogenesis. As a constitute of the hypothalamic-pituitary–gonadal axis, Sertoli cells promote spermatogenesis via protecting, nourishing, and supporting germ cells upon hormone determination. Here we clarified how the hormones in the hypothalamic-pituitary–gonadal axis, including FSH, testosterone and LH, regulate spermatogenesis via the androgen receptor, cAMP/PKA, PI3k/Akt signaling pathways in Sertoli cells. Other endogenous hormones in higher vertebrates, including ouabain, estradiol, leptin, MIS, PGD2, and thyroid hormone, also regulate spermatogenesis via the AR or cAMP/PKA signaling pathway. Among them, the dynamics of adherens junctions, gap junctions, and blood–testis barrier, glucose uptake, lactate supply and differentiation of Sertoli cells are regulated by more comprehensive hormones and signaling pathways in Sertoli cells. In infertile patients or patients with blocked spermatogenesis, the AR, cAMP/PKA and PI3k/Akt signaling pathways and related components exhibit abnormal activity or disordered content. The clinical specimens from patients with testicular cancer show similar mutated AR genes. According to the existing clinical evidence, it is valuable to study the deep mechanism of male infertility and testicular tumors from the perspective of hormones and signaling pathways in Sertoli cells.

Introduction

Cell differentiation happens when a cell changes into another specific cell type. It is a progressive and coordinated process which is regulated by signaling pathways (Sun et al., 2015a, Sun et al., 2015b). Spermatogenesis is an essential differentiation process for male reproduction in vertebrates (Wong and Cheng, 2005). Once spermatogenesis is disturbed, male infertility and even tumor will develop which will interfere with male health.

Spermatogenesis in higher vertebrates requires hormone regulation along the hypothalamic-pituitary–gonadal (HPG) axis, and disruption of HPG axis will impair sperm quality (Ilgin, 2020). Gonadotropin-releasing hormone (GnRH) is synthesized in the preoptic area of the hypothalamus and released to the anterior pituitary, which can stimulate the synthesis of follicle-stimulating hormone (FSH) and luteinising hormone (LH) (Abreu and Kaiser, 2016). FSH and LH regulate spermatogenesis and steroidogenesis in testis at puberty (for reviews, see Bhattacharya et al., 2019a, Bhattacharya et al., 2019b, Coss, 2020), and also participate in the feedback regulation along the HPG axis (Kanda, 2018). In the context of the HPG axis, Sertoli cells (SCs) serve as protectors, supporters, and nourishers of spermatogenesis (Mäkelä et al., 2019). We’d like to pointed out that hormones play critical roles in this process, including androgen, estradiol, FSH, leptin, LH, Müllerian-inhibiting substance (MIS), ouabain, prostaglandin D2 (PGD2), thyroid hormone, etc. (Fig. 1).

As the only somatic cells in the seminiferous epithelium in the testis, SCs are entwined with spermatogenic cells via cell junctions. SCs secret hormone, lactate and cytokines to promote mitosis, meiosis, and maturation of germ cells (GCs) (Morohoshi et al., 2019). Between SCs, there are cell junctions forming the blood–testis barrier (BTB), which is a physical protective barrier between the basal and the adluminal compartments of the seminiferous epithelium. Generally, only the preleptotene and leptotene spermatocytes are allowed to cross the BTB into the adluminal compartment to continue spermatogenesis (Ahmed et al., 2018). Disturbing any function of SCs can contribute to the inability of spermatogenesis. The failure organization of cell junction or inadequate supply of growth factors by SCs are recognized as the primary causes of Sertoli cell only syndrome (SCOS) (Paduch et al., 2019). In patients with non-obstructive azoospermia (NOA), disruption of BTB and adherens junction (AJ) between SCs and GCs are also observed (Zhu et al., 2019).

Owe to their precise regulation, the signaling pathways in SCs are vital for spermatogenesis (Wang et al., 2019a, Wang et al., 2019b). We had reviewed the mitogen-activated protein kinase (MAPK), adenosine 5‘-monophosphate (AMP)-activated protein kinase (AMPK), and transforming growth factor-β/Sma- and Mad-related protein (TGF-β/Smad) signaling pathways in spermatogenesis. Those three pathways regulate spermatogenesis via SCs proliferation, cell junction dynamics, lactate supply, self-renewal of spermatogonial stem cells (SSCs), and apoptosis of GCs as well (Ni et al., 2019, Pradhan et al., 2019). However, the spermatogenesis supporting functions of SCs are not restricted in their determination by the intracellular signaling pathways, but also regulated by hormones. Meroni et al. had reviewed that endogenic hormones including FSH can participate in the proliferation of SCs via the cyclic adenosine monophosphate/cAMP-dependent protein kinase (cAMP/PKA), extracellular regulated protein kinase 1/2 (ERK1/2), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt), and mechanistic target of rapamycin kinase complex 1 (mTORC1) signaling pathways (Meroni et al., 2019). We surveyed articles from 1982 to 2020 in the Pubmed database with following search terms: signaling pathway*, Sertoli cell*, spermatogenesis, hormone*, AR, androgen receptor, cAMP, PKA, PI3k, Akt, testosterone, androgen, ouabain, MIS, estradiol, LH, FSH, thyroid hormone, prostaglandin, leptin, male infertility, testicular cancer*, SCOS, NOA. We choose the findings which study how signaling pathways in SCs regulate spermatogenesis upon hormone regulation. Relevant articles about certain genes are also included as an extension or supplement. According to our investigation, hormones along the HPG axis and other endogenous hormones can be critical for spermatogenesis of higher vertebrates via their regulation of signaling pathways in SCs. The androgen receptor (AR), cAMP/PKA and PI3k/Akt signaling pathways are significant in this physiological regulation process (Table 1). In our review, we try to clarify this issue from the organ/system perspective. For contradictory outcomes of clinical trials, we put forward our analyses and suggestions to improve further studies. Based on the abnormal activity of signaling pathways and levels of hormone in clinical specimens, we also discuss possible clinical applications and productive approaches to clarify pathogenesis mechanisms, and develop therapies for testicular cancer and male infertility.

Section snippets

The AR signaling pathway

LH from pituitary can stimulate the Leydig cells in the seminiferous tubules to produce androgens. After androgens diffuse to SCs and bind to AR, the AR signaling pathway is initiated (Smith and Walker, 2014). Testosterone is the androgen in the testis, whose receptor expresses in SCs (Walker, 2011), so the AR signaling pathway in SCs are crucial for androgen mediated spermatogenesis (Soffientini et al., 2017) (Fig. 2).

The AR signaling pathway includes classical and non-classical testosterone

The cAMP/PKA signaling pathway

The de novo activation of the cAMP/PKA signaling pathway is tightly and directly connected to FSH (Eto et al., 2012). During the first spermatogenic wave in mice from 8 dpp onward, increasing FSH binds to its receptor, which is then coupled to stimulatory G-proteins complex (Gs)-coupled seven-transmembrane receptor (Simoni et al., 1997){Simoni, 1997 #61}. Gsα dissociates from FSH receptor and activates adenylyl cyclase (AC). Then the intracellular cAMP level is increased upon the catalysis of

The PI3k/Akt signaling pathway

FSH, insulin and insulin-like growth factor (IGF) highly participate in the PI3k/Akt signaling pathway (Cannarella et al., 2018) (Fig. 4). When IGF and insulin bind to IGF receptor (IGFR), firstly insulin receptor substrate 1 (IRS1) and then PI3K are phosphorylated (Mok et al., 2014). The phosphorylated PI3k converts phosphatidylinositol-4,5-bisphosphate (PIP2) lipids to phosphatidylinositol-3,4,5-trisphosphate (PIP3). Then PIP3 activates a subsequent phosphorylation cascade of

The abnormal spermatogenesis: Infertility

Infertility is a worldwide health issue, disturbing individuals, families, society and countries for a long history. Among them, 15% of the infertile men were diagnosed as azoospermia, whose ejaculate lack sperm according to the clinical manifestation (Wosnitzer and Goldstein, 2016). Azoospermia is classified into obstructive azoospermia and NOA, and SCOS is diagnosed in 26.3%-57.8% of azoospermic patients (Abofoul-Azab et al., 2019). Benefiting from the progression in microsurgical vasal

Abnormal spermatogenesis: Testicular tumor

Testicular cancer is the most common type of cancer in men aged 15–34 years old. In 2016, 8700 cases of testicular cancer and 380 deaths were estimated in the United States (Smith et al., 2018). While in 2017, the corresponding indexes turn into 8850 and 410 (Baird et al., 2018). Sertoli cell tumor (SCT) is a kind of sex-cord stromal tumor, and only accounts for 0.4%-1.5% of testicular malignancies (Coleman et al., 2014, Ishida et al., 2013). Existing cases indicate the relevance between the

Conclusions and perspectives

Currently, studies are rarely focusing on the relationship of hormone determined signaling pathways in SCs, spermatogenesis, and clinical cases. However, the signaling pathways, especially the AR signaling pathway, promise a great future in diagnosis and therapy of male diseases, as well as drug development. Mutation in the AR exon and expression level of AR in SCs, as well as downstream genes of the AR signaling pathway can serve as markers of diseases to screen potential therapies and agents

Funding Information

This work was supported in part by National Natural Science Foundation of China (No 41776144) and Zhejiang Province Public Welfare Technology Application Research Project (including Natural Science Foundation) (No. LGF20C120001).

Author Contribution

F.-D.N, S.-L.H., and W.-X.Y. conceived of and authored the paper.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

The authors are grateful to all members of the Sperm Laboratory at Zhejiang University for valuable discussions.

References (164)

  • J. Don et al.

    The expanding family of CREB/CREM transcription factors that are involved with spermatogenesis

    Mol. Cell. Endocrinol.

    (2002)
  • K. Eto et al.

    Nociceptin is upregulated by FSH signaling in Sertoli cells in murine testes

    Biochem. Biophys. Res. Commun.

    (2012)
  • K. Eto

    Nociceptin and meiosis during spermatogenesis in postnatal testes

    Vitam. Horm.

    (2015)
  • E. Fynn-Thompson et al.

    Inhibition of steroidogenesis in Leydig cells by Müllerian-inhibiting substance

    Mol. Cell. Endocrinol.

    (2003)
  • Y. Hai et al.

    The roles and regulation of Sertoli cells in fate determinations of spermatogonial stem cells and spermatogenesis

    Semin. Cell Dev. Biol.

    (2014)
  • M.P. Hedger et al.

    Regulation of activin and inhibin in the adult testis and the evidence for functional roles in spermatogenesis and immunoregulation

    Mol. Cell Endocrinol.

    (2012)
  • S. Hongo et al.

    Induction of asparagine synthetase by follicle-stimulating hormone in primary cultures of rat Sertoli cells

    Arch. Biochem. Biophys.

    (1994)
  • I. Knoke et al.

    A(870)E mutation of the androgen receptor gene in a patient with complete androgen insensitivity syndrome and Sertoli cell tumor

    Cancer Genet. Cytogenet.

    (1997)
  • L. Konrad et al.

    Cardiotonic steroids trigger non-classical testosterone signaling in Sertoli cells via the α4 isoform of the sodium pump

    Biochim. Biophys. Acta.

    (2011)
  • A. Kumar et al.

    Direct regulation of genes involved in sperm release by estrogen and androgen through their receptors and coregulators

    J. Steroid. Biochem. Mol. Biol.

    (2017)
  • N. Kumar et al.

    Assessment of estradiol-induced gene regulation and proliferation in an immortalized mouse immature Sertoli cell line

    Life Sci.

    (2016)
  • D. Lamberti et al.

    Promoter analysis of the gene encoding GDNF in murine Sertoli cells

    Mol. Cell. Endocrinol.

    (2014)
  • W.W. Lin et al.

    Absence of cyclic adenosine 3':5' monophosphate responsive element modulator expression at the spermatocyte arrest stage

    Fertil. Steril.

    (1998)
  • M.E. Matzkin et al.

    Exploring the cyclooxygenase 2 (COX2)/15d-△12,14PGJ2 system in hamster Sertoli cells: regulation by FSH/testosterone and relevance to glucose uptake

    Gen. Comp. Endocrinol.

    (2012)
  • A. Morohoshi et al.

    The ubiquitin ligase subunit β-TrCP in Sertoli cells is essential for spermatogenesis in mice

    Dev. Biol.

    (2019)
  • A.R. Nascimento et al.

    Intracellular signaling pathways involved in the relaxin-induced proliferation of rat Sertoli cells

    Eur. J. Pharmacol.

    (2012)
  • M. Abofoul-Azab et al.

    Identification of Premeiotic, Meiotic, and Postmeiotic Cells in Testicular Biopsies Without Sperm from Sertoli Cell-Only Syndrome Patients

    Int. J. Mol. Sci.

    (2019)
  • AuthorsRamphul, K., Zulfiqar, H., Mejias, S.G., 2020. Sertoli-Cell-Only Syndrome. Treasure Island (FL): StatPearls...
  • D.C. Baird et al.

    Testicular Cancer: Diagnosis and Treatment

    Am. Fam. Physician

    (2018)
  • W.A. Banks et al.

    Passage of leptin across the blood-testis barrier

    Am. J. Physiol.

    (1999)
  • I. Bhattacharya et al.

    Low levels of Gαs and Ric8b in testicular sertoli cells may underlie restricted FSH action during infancy in primates

    Endocrinology

    (2015)
  • I. Bhattacharya et al.

    Pubertal orchestration of hormones and testis in primates

    Mol. Reprod. Dev.

    (2019)
  • C. Bois et al.

    Age- and cell-related gene expression of aromatase and estrogen receptors in the rat testis

    J Mol. Endocrinol.

    (2010)
  • B.P. Borjian et al.

    Study of trinucleotide expansions and expression of androgen receptor in infertile men with abnormal spermogram referred to Royan institute

    Andrologia

    (2018)
  • L. Braydich-Stolle et al.

    Role of glial cell line-derived neurotrophic factor in germ-line stem cell fate

    Ann. N. Y. Acad. Sci.

    (2005)
  • J.J. Buzzard et al.

    Thyroid hormone, retinoic acid, and testosterone suppress proliferation and induce markers of differentiation in cultured rat sertoli cells

    Endocrinology

    (2003)
  • K. Cai et al.

    Action mechanism of inhibin α-subunit on the development of Sertoli cells and first wave of spermatogenesis in mice

    PLoS ONE

    (2011)
  • R. Cannarella et al.

    Effects of the insulin-like growth factor system on testicular differentiation and function: a review of the literature

    Andrology

    (2018)
  • L. Casarini et al.

    FSH for the Treatment of Male Infertility

    Int. J. Mol. Sci.

    (2020)
  • E. Castro-Nallar et al.

    Androgen receptor gene CAG and GGN repeat polymorphisms in Chilean men with primary severe spermatogenic failure

    J. Androl.

    (2010)
  • Cavalcante-Silva, L.H.A, Lima, E.A., Carvalho, D.C.M., de, Sales-Neto, J.M., Alves, A.K.A., Galvao, J., da, Silva,...
  • S.R. Chen et al.

    Androgen receptor in Sertoli cells regulates DNA double-strand break repair and chromosomal synapsis of spermatocytes partially through intercellular EGF-EGFR signaling

    Oncotarget

    (2016)
  • J. Cheng et al.

    Testosterone activates mitogen-activated protein kinase via Src kinase and the epidermal growth factor receptor in sertoli cells

    Endocrinology

    (2007)
  • V.L. Chin et al.

    A case of complete androgen insensitivity syndrome with a novel androgen receptor mutation

    J. Pediatr. Endocrinol. Metab.

    (2012)
  • S. Coleman et al.

    Testicular sclerosing sertoli cell tumor: a case report and review of the literature

    Rev. Urol.

    (2014)
  • R.J. Coombs et al.

    Only tissue-type plasminogen activator is secreted by immature bovine Sertoli cell-enriched cultures

    J. Endocrinol.

    (1988)
  • D. Coss

    Commentary on the Recent FSH Collection: Known Knowns and Known Unknowns

    Endocrinology

    (2020)
  • P. Crepieux et al.

    The ERK-dependent signalling is stage-specifically modulated by FSH, during primary Sertoli cell maturation

    Oncogene

    (2001)
  • C. Daems et al.

    MEF2 Cooperates With Forskolin/cAMP and GATA4 to Regulate Star Gene Expression in Mouse MA-10 Leydig Cells

    Endocrinology

    (2015)
  • F. Delfino et al.

    Stage-specific nuclear expression of NF-κB in mammalian testis

    Mol. Endocrinol.

    (1998)
  • Cited by (0)

    1

    Authors contributed equally to this work.

    View full text