Skip to main content

Testicular Signaling: Team Work in Sperm Production

  • Chapter
  • First Online:

Abstract

The male gonads, testis, have two main functions: testosterone production (steroidogenesis), a fundamental hormone for the development and maintenance of several physiological functions; and sperm production (spermatogenesis), essential for male fertility. The synthesis of both products is mainly regulated by endocrine hormones, synthesized in the hypothalamus and pituitary gland, and paracrine signals. This chapter will explore the signaling pathways involved in testosterone production by Leydig cells. We will also discuss both classical and non-classical pathways of testosterone action in spermatogenesis, and the contribution of follicle stimulating hormone to spermatogenesis maintenance. Finally, the signaling pathways involved in blood-testis-barrier regulation as well as other paracrine signals involved in spermatogenesis control will be explored. Despite these pathways occur in most somatic cells, they have a unique role in regulating the most peculiar and exceptional process in one of the most complex tissue in male body.

J. Santiago and D. Patrício—Contributed equally.

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   99.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD   129.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

Abbreviations

17OH-Allo:

17OH-allopregnanolone

17OH-DHP:

17OH-dihydroprogesterone

17OHPreg:

17α-hydroxypregnenolone

17OHProg:

17α-hydroxyprogesterone

AA:

Arachidonic acid

AC:

Adenylyl cyclase

AKR1C2/4:

Aldo-keto reductase family 1 member C2/4

AKR1C3:

Aldo-keto reductase family 1 member C3

AKT:

RAC-alpha serine/threonine-protein kinase

AKT1S1:

Proline-rich AKT1 substrate 1

AR:

Androgen receptor

ARE:

Androgen response elements

ATP:

Adenosine triphosphate

BCL6B:

B cell CLL/lymphoma 6 member B

BTB:

Blood-Testis-Barrier

CAM:

Calmodulin

cAMP:

Cyclic adenosine monophosphate

CAR:

Coxsackievirus and adenovirus receptor

CDC42:

Cell division control protein 42 homolog

CDK1:

Cyclin D1

CLDN:

Claudin

CRE:

cAMP-response element

CREB:

Cyclic AMP-responsive element-binding protein 1

CREM:

cAMP-responsive element modulators

CSF1:

Colony stimulating factor

CXCL12:

Stromal cell-derived factor 1

CXCR4:

C-X-C chemokine receptor type 4

CYP11A1:

Cholesterol side-chain cleavage enzyme

CYP17A1:

Steroid 17-alpha-hydroxylase/17,20 lyase

CYP19A1:

Aromatase

DAG:

Diacylglycerol

DHEA:

Dehydroepiandrosterone

DHT:

Dihydrotestosterone

DMRT:

Double sex- and mab-3 related transcription factor

DNA:

Deoxyribonucleic acid

EGF:

Epidermal growth factor

EGFR:

Epidermal growth factor receptor

EGR1:

Early growth response protein 1

ER:

Endoplasmic reticulum

ERBB4:

Receptor tyrosine-protein kinase erbB-4

ERK:

Mitogen-activated protein kinase

ES:

Ectoplasmic specialization

ETV5:

ETS translocation variant 5

FAK:

Focal adhesion kinases

FDX1:

Ferredoxin-1

FDXR:

Ferredoxin reductase

FGF:

Fibroblast growth factor

FSH:

Follicle stimulating hormone

FSHR:

Follicle stimulating hormone receptor

GATA-4:

Transcription factor GATA-4

GDF9:

Growth/differentiation factor 9

GDNF:

Glial cell-derived neurotrophic factor

GFRA1:

GDNF family receptor alpha-1

GNE:

Guanine nucleotide exchange factor

GnRH:

Gonadotropin releasing hormone

GPRC:

G protein-coupled receptor

HSD17B3:

17β-hydroxysteroid dehydrogenase type 3

HSD3B2:

3β-hydroxysteroid dehydrogenase type II

HSP:

Heat shock proteins

ID4:

DNA-binding protein 4

IGF:

Insulin-like growth factor

IL:

Interleukins

IMM:

Inner mitochondrial membrane

INSL3:

Insulin-like factor 3

IP3:

Inositol 1,4,5-trisphosphate

JAM:

Junctional adhesion molecules

KITLG:

Stem cell factor/kit ligand

KO:

Knock out

LC:

Leydig Cell

LDH:

Lactate dehydrogenase

LDHA:

L-lactate dehydrogenase A chain

LH:

Luteinizing Hormone

LHCGR:

Luteinizing hormone/choriogonadotropin receptor

LHX1:

LIM homeobox 1

LICH:

Cholesteryl ester hydrolase

MAPK:

Mitogen-activated protein kinase

MMP9:

Matrix metalloproteinase-9

mTOR:

Serine/threonine-protein kinase mTOR

mTORC1:

Mammalian target of rapamycin complex 1

mTORC2:

Mammalian target of rapamycin complex 2

NF-κB:

Nuclear factor-kappa B

NRG:

Neuregulin

OCLN:

Ocludin

OPRL1:

Nociceptin receptor

PDPK1:

Phosphoinositide dependent protein kinase 1

PDE:

Phosphodiesterase

PGE2:

Prostaglandin E2

PIK3:

Phosphatidylinositol 3 kinase

PIP2:

Phosphatidylinositol 4,5-bisphosphate

PLA2:

Phospholipase A2

PLC:

Phospholipase C

POR:

P450 oxidoreductase

PRKA:

Protein kinase A

PRKB:

Protein kinase B

PRKC:

Protein kinase C

RA:

Retinoic acid

RAF:

RAF proto-oncogene serine/threonine-protein kinase

RAR:

Nuclear retinoic receptor

REC8:

Meiotic recombination protein rec8

RHOX5:

Homeobox protein Rhox5

RNA:

Ribonucleic acid

RPS6K:

Ribosomal protein S6 kinase

RoDH:

Retinol dehydrogenase

RXR:

Retinoic X receptor

SC:

Sertoli Cell

SCAR:

Sertoli cells-specific androgen receptor

SFK:

SRC family kinase

SRC:

Proto-oncogene tyrosine-protein kinase Src

SRD5A2:

5α-reductase type II

SRF:

Serum response factor

SSC:

Spermatogonial stem cells

StAR:

Steroidogenic acute regulatory protein

STF1:

Steroidogenic factor 1

TGF:

Transforming growth factor

TJ:

Thigh junctions

TNF:

Tumor necrosis factor

VEGF:

Vascular endothelial growth factor

VIM:

Vimentin

γ-GTP:

γ-glutamyl peptidase

References

  1. Kerr JB (1992) Functional cytology of the human testis. Baillieres Clin Endocrinol Metab 6:235–250. https://doi.org/10.1016/S0950-351X(05)80149-1

    Article  CAS  PubMed  Google Scholar 

  2. Moghimian M, Soltani M, Abtahi H et al (2016) Protective effect of tunica albuginea incision with tunica vaginalis flap coverage on tissue damage and oxidative stress following testicular torsion: role of duration of ischemia. J Pediatr Urol 12:390.e1–390.e6. https://doi.org/10.1016/j.jpurol.2016.06.002

    Article  CAS  PubMed  Google Scholar 

  3. de Kretser DM, Loveland KL, Meinhardt A et al (1998) Spermatogenesis. Hum Reprod 13:1–8. https://doi.org/10.1093/humrep/13.suppl_1.1

    Article  PubMed  Google Scholar 

  4. Johnson L, Thompson DL, Varner DD (2008) Role of Sertoli cell number and function on regulation of spermatogenesis. Anim Reprod Sci 105:23–51. https://doi.org/10.1016/j.anireprosci.2007.11.029

    Article  CAS  PubMed  Google Scholar 

  5. Ferlin A, Arredi B, Zuccarello D et al (2006) Paracrine and endocrine roles of insulin-like factor 3. J Endocrinol Invest 29:657–664. https://doi.org/10.1007/BF03344168

    Article  CAS  PubMed  Google Scholar 

  6. Haider SG (2004) Cell biology of Leydig cells in the testis. Int Rev Cytol 233:181–241. https://doi.org/10.1016/S0074-7696(04)33005-6

    Article  CAS  PubMed  Google Scholar 

  7. Rajender S, Rahul P, Mahdi AA (2010) Mitochondria, spermatogenesis and male infertility. Mitochondrion 10:419–428. https://doi.org/10.1016/j.mito.2010.05.015

    Article  CAS  PubMed  Google Scholar 

  8. Smith LB, Walker WH (2014) The regulation of spermatogenesis by androgens. Semin Cell Dev Biol 30:2–13. https://doi.org/10.1016/j.semcdb.2014.02.012

    Article  CAS  PubMed  Google Scholar 

  9. Luisi S, Florio P, Reis FM, Petraglia F (2005) Inhibins in female and male reproductive physiology: role in gametogenesis, conception, implantation and early pregnancy. Hum Reprod Update 11:123–135. https://doi.org/10.1093/humupd/dmh057

    Article  CAS  PubMed  Google Scholar 

  10. Andreone L, Ambao V, Pellizzari EH et al (2017) Role of FSH glycan structure in the regulation of Sertoli cell inhibin production. Reproduction 154:711–721. https://doi.org/10.1530/REP-17-0393

    Article  CAS  PubMed  Google Scholar 

  11. Bronson R (2011) Biology of the male reproductive tract: its cellular and morphological considerations. Am J Reprod Immunol 65:212–219. https://doi.org/10.1111/j.1600-0897.2010.00944.x

    Article  CAS  PubMed  Google Scholar 

  12. Tilbrook AJ, Clarke IJ (2001) Negative feedback regulation of the secretion and actions of gonadotropin-releasing hormone in males. Biol Reprod 64:735–742. https://doi.org/10.1095/biolreprod64.3.735

    Article  CAS  PubMed  Google Scholar 

  13. Barsoum IB, Yao HH-C (2010) Fetal Leydig cells: progenitor cell maintenance and differentiation. J Androl 31:11–15. https://doi.org/10.2164/jandrol.109.008318

    Article  CAS  PubMed  Google Scholar 

  14. Benton L, Shan LX, Hardy MP (1995) Differentiation of adult Leydig cells. J Steroid Biochem Mol Biol 53:61–68. https://doi.org/10.1016/0960-0760(95)00022-R

    Article  CAS  PubMed  Google Scholar 

  15. Stocco DM, Wang X, Jo Y, Manna PR (2005) Multiple signaling pathways regulating steroidogenesis and steroidogenic acute regulatory protein expression: more complicated than we thought. Mol Endocrinol 19:2647–2659. https://doi.org/10.1210/me.2004-0532

    Article  CAS  PubMed  Google Scholar 

  16. Tremblay JJ (2015) Molecular regulation of steroidogenesis in endocrine Leydig cells. Steroids 103:3–10. https://doi.org/10.1016/j.steroids.2015.08.001

    Article  CAS  PubMed  Google Scholar 

  17. Miller WL, Auchus RJ (2011) The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev 32:81–151. https://doi.org/10.1210/er.2010-0013

    Article  PubMed  Google Scholar 

  18. Selvaraj ​V, Stocco DM, Clark BJ (2018) Current knowledge on the acute regulation of steroidogenesis. Biol Reprod 99:13–26. https://doi.org/10.1093/biolre/ioy102

  19. Marti N, Galván JA, Pandey AV et al (2017) Genes and proteins of the alternative steroid backdoor pathway for dihydrotestosterone synthesis are expressed in the human ovary and seem enhanced in the polycystic ovary syndrome. Mol Cell Endocrinol 441:116–123. https://doi.org/10.1016/j.mce.2016.07.029

    Article  CAS  PubMed  Google Scholar 

  20. Auchus RJ (2004) The backdoor pathway to dihydrotestosterone. Trends Endocrinol Metab 15:432–438. https://doi.org/10.1016/j.tem.2004.09.004

    Article  CAS  PubMed  Google Scholar 

  21. Fukami M, Homma K, Hasegawa T, Ogata T (2013) Backdoor pathway for dihydrotestosterone biosynthesis: implications for normal and abnormal human sex development. Dev Dyn 242:320–329. https://doi.org/10.1002/dvdy.23892

    Article  CAS  PubMed  Google Scholar 

  22. Flück CE, Meyer-Böni M, Pandey AV et al (2011) Why boys will be boys: two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation. Am J Hum Genet. https://doi.org/10.1016/j.ajhg.2011.06.009

    Article  PubMed  PubMed Central  Google Scholar 

  23. Oliveira PF, Alves MG (2015) Sertoli cell and germ cell differentiation. In: Sertoli cell metabolism and spermatogenesis. Springer International Publishing, Cham, pp 25–39

    Google Scholar 

  24. Walker WH (2010) Non-classical actions of testosterone and spermatogenesis. Philos Trans R Soc B Biol Sci 365:1557–1569. https://doi.org/10.1098/rstb.2009.0258

    Article  CAS  Google Scholar 

  25. Mruk DD, Cheng CY (2015) The mammalian blood-testis barrier: its biology and regulation. Endocr Rev 36:564–591. https://doi.org/10.1210/er.2014-1101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Walker WH, Cheng J (2005) FSH and testosterone signaling in Sertoli cells. Reproduction 130:15–28. https://doi.org/10.1530/rep.1.00358

    Article  CAS  Google Scholar 

  27. Rebourcet D, Darbey A, Monteiro A et al (2017) Sertoli cell number defines and predicts germ and leydig cell population sizes in the adult mouse testis. Endocrinology 158:2955–2969. https://doi.org/10.1210/en.2017-00196

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gallay N, Gagniac L, Guillou F, Crépieux P (2014) The follicle-stimulating hormone signaling network in Sertoli cells. In: Cellular endocrinology in health and disease. Elsevier, pp 85–100

    Google Scholar 

  29. Kumar TR, Wang Y, Lu N, Matzuk MM (1997) Follicle stimulating hormone is required for ovarian follicle maturation but not male fertility. Nat Genet 15:201–204. https://doi.org/10.1038/ng0297-201

    Article  CAS  PubMed  Google Scholar 

  30. Dierich A, Sairam MR, Monaco L et al (1998) 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 95:13612–13617. https://doi.org/10.1073/pnas.95.23.13612

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Abel MH, Wootton AN, Wilkins V et al (2000) The effect of a null mutation in the follicle-stimulating hormone receptor gene on mouse reproduction. Endocrinology 141:1795–1803. https://doi.org/10.1210/endo.141.5.7456

    Article  CAS  PubMed  Google Scholar 

  32. Krishnamurthy H, Babu PS, Morales CR, Sairam MR (2001) Delay in sexual maturity of the follicle-stimulating hormone receptor knockout male mouse. Biol Reprod 65:522–531. https://doi.org/10.1095/biolreprod65.2.522

    Article  CAS  PubMed  Google Scholar 

  33. Sairam MR, Krishnamurthy H (2001) The role of follicle-stimulating hormone in spermatogenesis: lessons from knockout animal models. Arch Med Res 32:601–608

    Article  CAS  PubMed  Google Scholar 

  34. Kangasniemi M, Kaipia A, Mali P et al (1990) Modulation of basal and FSH-dependent cyclic AMP production in rat seminiferous tubules staged by an improved transillumination technique. Anat Rec 227:62–76. https://doi.org/10.1002/ar.1092270108

    Article  CAS  PubMed  Google Scholar 

  35. Pitetti J-L, Calvel P, Zimmermann C et al (2013) An essential role for insulin and IGF1 receptors in regulating sertoli cell proliferation, testis size, and FSH action in mice. Mol Endocrinol 27:814–827. https://doi.org/10.1210/me.2012-1258

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Crépieux P, Marion S, Martinat N et al (2001) The ERK-dependent signalling is stage-specifically modulated by FSH, during primary Sertoli cell maturation. Oncogene 20:4696–4709. https://doi.org/10.1038/sj.onc.1204632

    Article  PubMed  Google Scholar 

  37. Nascimento AR, Macheroni C, Lucas TFG et al (2016) Crosstalk between FSH and relaxin at the end of the proliferative stage of rat Sertoli cells. Reproduction 152:613–628. https://doi.org/10.1530/REP-16-0330

    Article  CAS  PubMed  Google Scholar 

  38. Huhtaniemi I (2015) A short evolutionary history of FSH-stimulated spermatogenesis. Hormones 14:468–478. https://doi.org/10.14310/horm.2002.1632

  39. Walker WH, Fucci L, Habener JF (1995) Expression of the gene encoding transcription factor cyclic adenosine 3′,5′-monophosphate (cAMP) response element-binding protein (CREB): regulation by follicle-stimulating hormone-induced cAMP signaling in primary rat Sertoli cells. Endocrinology 136:3534–3545. https://doi.org/10.1210/endo.136.8.7628390

    Article  CAS  PubMed  Google Scholar 

  40. Scobey MJ, Bertera S, Somers JP et al (2001) Delivery of a cyclic adenosine 3′,5′-monophosphate response element-binding protein (CREB) mutant to seminiferous tubules results in impaired spermatogenesis. Endocrinology 142:948–954. https://doi.org/10.1210/endo.142.2.7948

    Article  CAS  PubMed  Google Scholar 

  41. Foulkes NS, Schlotter F, Pévet P, Sassone-Corsi P (1993) Pituitary hormone FSH directs the CREM functional switch during spermatogenesis. Nature 362:264–267. https://doi.org/10.1038/362264a0

    Article  CAS  PubMed  Google Scholar 

  42. Nantel F, Monaco L, Foulkes NS et al (1996) Spermiogenesis deficiency and germ-cell apoptosis in CREM-mutant mice. Nature 380:159–162. https://doi.org/10.1038/380159a0

    Article  CAS  PubMed  Google Scholar 

  43. Ruwanpura SM, McLachlan RI, Meachem SJ (2010) Hormonal regulation of male germ cell development. J Endocrinol 205:117–131. https://doi.org/10.1677/JOE-10-0025

    Article  CAS  PubMed  Google Scholar 

  44. Franchi E, Camatini M (1985) Evidence that a Ca2+ chelator and a calmodulin blocker interfere with the structure of inter-sertoli junctions. Tissue Cell 17:13–25. https://doi.org/10.1016/0040-8166(85)90012-6

    Article  CAS  PubMed  Google Scholar 

  45. Grasso P, Reichert LE (1989) Follicle-stimulating hormone receptor-mediated uptake of 45Ca2+ by proteoliposomes and cultured rat sertoli cells: evidence for involvement of voltage-activated and voltage-independent calcium channels. Endocrinology 125:3029–3036. https://doi.org/10.1210/endo-125-6-3029

    Article  CAS  PubMed  Google Scholar 

  46. Gorczynska E, Handelsman DJ (1991) The role of calcium in follicle-stimulating hormone signal transduction in Sertoli cells. J Biol Chem 266:23739–23744. https://doi.org/10.1210/endo.134.4.8137759

    Article  CAS  PubMed  Google Scholar 

  47. Lalevée N, Pluciennik F, Joffre M (1997) Voltage-dependent calcium current with properties of T-type current in Sertoli cells from immature rat testis in primary cultures. Biol Reprod 56:680–687. https://doi.org/10.1095/biolreprod56.3.680

    Article  PubMed  Google Scholar 

  48. Wu G-Y, Deisseroth K, Tsien RW (2001) Activity-dependent CREB phosphorylation: convergence of a fast, sensitive calmodulin kinase pathway and a slow, less sensitive mitogen-activated protein kinase pathway. Proc Natl Acad Sci 98:2808–2813. https://doi.org/10.1073/pnas.051634198

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Meroni SB, Riera MF, Pellizzari EH, Cigorraga SB (2002) Regulation of rat Sertoli cell function by FSH: possible role of phosphatidylinositol 3-kinase/protein kinase B pathway. J Endocrinol 174:195–204. https://doi.org/10.1677/joe.0.1740195

    Article  CAS  PubMed  Google Scholar 

  50. Riera MF, Regueira M, Galardo MN et al (2012) Signal transduction pathways in FSH regulation of rat Sertoli cell proliferation. Am J Physiol Metab 302:E914–E923. https://doi.org/10.1152/ajpendo.00477.2011

    Article  CAS  Google Scholar 

  51. Jannini EA, Ulisse S, Cecconi S et al (1994) Follicle-stimulating hormone-induced phospholipase A2 activity and eicosanoid generation in rat Sertoli cells. Biol Reprod 51:140–145. https://doi.org/10.1095/biolreprod51.1.140

    Article  CAS  PubMed  Google Scholar 

  52. Verhoeven G, Cailleau J (1988) Follicle-stimulating hormone and androgens increase the concentration of the androgen receptor in Sertoli cells. Endocrinology 122:1541–1550. https://doi.org/10.1210/endo-122-4-1541

    Article  CAS  PubMed  Google Scholar 

  53. Blok LJ, Mackenbach P, Trapman J et al (1989) Follicle-stimulating hormone regulates androgen receptor mRNA in Sertoli cells. Mol Cell Endocrinol 63:267–271. https://doi.org/10.1016/0303-7207(89)90104-4

    Article  CAS  PubMed  Google Scholar 

  54. Sadate-Ngatchou PI, Pouchnik DJ, Griswold MD (2004) Follicle-stimulating hormone induced changes in gene expression of murine testis. Mol Endocrinol 18:2805–2816. https://doi.org/10.1210/me.2003-0203

    Article  CAS  PubMed  Google Scholar 

  55. Jarow JP, Chen H, Rosner W et al (2001) Assessment of the androgen environment within the human testis: minimally invasive method to obtain intratesticular fluid. J Androl 22:640–645. https://doi.org/10.1002/j.1939-4640.2001.tb02224.x

    Article  CAS  PubMed  Google Scholar 

  56. Zirkin BR, Santulli R, Awoniyi CA, Ewing LL (1989) Maintenance of advanced spermatogenic cells in the adult rat testis: quantitative relationship to testosterone concentration within the testis. Endocrinology 124:3043–3049. https://doi.org/10.1210/endo-124-6-3043

    Article  CAS  PubMed  Google Scholar 

  57. Chang C, Chen Y-T, Yeh S-D et al (2004) Infertility with defective spermatogenesis and hypotestosteronemia in male mice lacking the androgen receptor in Sertoli cells. Proc Natl Acad Sci 101:6876–6881. https://doi.org/10.1073/pnas.0307306101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. De Gendt K, Swinnen JV, Saunders PTK et al (2004) A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci 101:1327–1332. https://doi.org/10.1073/pnas.0308114100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Verhoeven G, Willems A, Denolet E et al (2010) Androgens and spermatogenesis: lessons from transgenic mouse models. Philos Trans R Soc B Biol Sci 365:1537–1556. https://doi.org/10.1098/rstb.2009.0117

    Article  CAS  Google Scholar 

  60. Shiraishi K, Matsuyama H (2017) Gonadotoropin actions on spermatogenesis and hormonal therapies for spermatogenic disorders [Review]. Endocr J 64:123–131. https://doi.org/10.1507/endocrj.EJ17-0001

    Article  CAS  PubMed  Google Scholar 

  61. Walker WH (2011) Testosterone signaling and the regulation of spermatogenesis. Spermatogenesis 1:116–120. https://doi.org/10.4161/spmg.1.2.16956

    Article  PubMed  PubMed Central  Google Scholar 

  62. O’Hara L, Smith LB (2015) Androgen receptor roles in spermatogenesis and infertility. Best Pract Res Clin Endocrinol Metab 29:595–605. https://doi.org/10.1016/j.beem.2015.04.006

    Article  CAS  PubMed  Google Scholar 

  63. Lyon MF, Glenister PH, Lynn Lamoreux M (1975) Normal spermatozoa from androgen-resistant germ cells of chimaeric mice and the role of androgen in spermatogenesis. Nature 258:620–622. https://doi.org/10.1038/258620a0

    Article  CAS  PubMed  Google Scholar 

  64. Griswold MD (1998) The central role of Sertoli cells in spermatogenesis. Semin Cell Dev Biol 9:411–416. https://doi.org/10.1006/scdb.1998.0203

    Article  CAS  PubMed  Google Scholar 

  65. Meng J, Holdcraft RW, Shima JE et al (2005) Androgens regulate the permeability of the blood-testis barrier. Proc Natl Acad Sci 102:16696–16700. https://doi.org/10.1073/pnas.0506084102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Holdcraft RW, Braun RE (2004) Hormonal regulation of spermatogenesis. Int J Androl 27:335–342. https://doi.org/10.1111/j.1365-2605.2004.00502.x

    Article  CAS  PubMed  Google Scholar 

  67. Lindsey JS, Wilkinson MF (1996) Pem: a testosterone- and LH-regulated homeobox gene expressed in mouse Sertoli cells and epididymis. Dev Biol 179:471–484. https://doi.org/10.1006/dbio.1996.0276

    Article  CAS  PubMed  Google Scholar 

  68. Zhou J, Pan J, Eckardt S et al (2011) Nxf3 is expressed in Sertoli cells, but is dispensable for spermatogenesis. Mol Reprod Dev 78:241–249. https://doi.org/10.1002/mrd.21291

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Cheng J, Watkins SC, Walker WH (2007) Testosterone activates mitogen-activated protein kinase via Src kinase and the epidermal growth factor receptor in sertoli cells. Endocrinology 148:2066–2074. https://doi.org/10.1210/en.2006-1465

    Article  CAS  PubMed  Google Scholar 

  70. Fix C, Jordan C, Cano P, Walker WH (2004) Testosterone activates mitogen-activated protein kinase and the cAMP response element binding protein transcription factor in Sertoli cells. Proc Natl Acad Sci 101:10919–10924. https://doi.org/10.1073/pnas.0404278101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. O’Donnell L, Stanton P, de Kretser DM (2000) Endocrinology of the male reproductive system and spermatogenesis. MDText.com, Inc.

    Google Scholar 

  72. Zhang J, Wong CH, Xia W et al (2005) Regulation of Sertoli-germ cell adherens junction dynamics via changes in protein-protein interactions of the N-cadherin-β-catenin protein complex which are possibly mediated by c-Src and myotubularin-related protein 2: an in vivo study using an androgen. Endocrinology 146:1268–1284. https://doi.org/10.1210/en.2004-1194

    Article  CAS  PubMed  Google Scholar 

  73. Shupe J, Cheng J, Puri P et al (2011) Regulation of Sertoli-germ cell adhesion and sperm release by fsh and nonclassical testosterone signaling. Mol Endocrinol 25:238–252. https://doi.org/10.1210/me.2010-0030

    Article  CAS  PubMed  Google Scholar 

  74. Xia W, Wong CH, Lee NPY et al (2005) Disruption of Sertoli-germ cell adhesion function in the seminiferous epithelium of the rat testis can be limited to adherens junctions without affecting the blood-testis barrier integrity: an in vivo study using an androgen suppression model. J Cell Physiol 205:141–157. https://doi.org/10.1002/jcp.20377

    Article  CAS  PubMed  Google Scholar 

  75. Lyng FM, Jones GR, Rommerts FFG (2000) Rapid androgen actions on calcium signaling in rat Sertoli cells and two human prostatic cell lines: similar biphasic responses between 1 picomolar and 100 nanomolar concentrations. Biol Reprod 63:736–747. https://doi.org/10.1095/biolreprod63.3.736

    Article  CAS  PubMed  Google Scholar 

  76. Gorczynska E, Handelsman DJ (1995) Androgens rapidly increase the cytosolic calcium concentration in Sertoli cells. Endocrinology 136:2052–2059. https://doi.org/10.1210/endo.136.5.7720654

    Article  CAS  PubMed  Google Scholar 

  77. Von Ledebur EICF, Almeida JP, Loss ES, Wassermann GF (2002) Rapid effect of testosterone on rat Sertoli cell membrane potential. Relationship with K+ ATP channels. Horm Metab Res 34:550–555. https://doi.org/10.1055/s-2002-35426

    Article  Google Scholar 

  78. Loss ES, Jacobsen M, Costa ZS et al (2004) Testosterone modulates K(+)ATP channels in Sertoli cell membrane via the PLC-PIP2 pathway. Horm Metab Res 36:519–525. https://doi.org/10.1055/s-2004-825753

    Article  CAS  PubMed  Google Scholar 

  79. Stanton PG (2016) Regulation of the blood-testis barrier. Semin Cell Dev Biol 59:166–173. https://doi.org/10.1016/j.semcdb.2016.06.018

    Article  CAS  PubMed  Google Scholar 

  80. Russell L (1977) Movement of spermatocytes from the basal to the adluminal compartment of the rat testis. Am J Anat 148:313–328. https://doi.org/10.1002/aja.1001480303

    Article  CAS  PubMed  Google Scholar 

  81. Dym M, Cavicchia JC (1977) Further observations on the blood-testis barrier in monkeys. Biol Reprod 17:390–403. https://doi.org/10.1095/biolreprod17.3.390

    Article  CAS  PubMed  Google Scholar 

  82. Pelletier R-M (2011) The blood-testis barrier: the junctional permeability, the proteins and the lipids. Prog Histochem Cytochem 46:49–127. https://doi.org/10.1016/j.proghi.2011.05.001

    Article  PubMed  Google Scholar 

  83. Hermo L, Pelletier R-M, Cyr DG, Smith CE (2010) Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 5: intercellular junctions and contacts between germs cells and Sertoli cells and their regulatory interactions, testicular cholesterol, and genes/proteins. Microsc Res Tech 73:409–494. https://doi.org/10.1002/jemt.20786

    Article  CAS  PubMed  Google Scholar 

  84. Mazaud-Guittot S, Meugnier E, Pesenti S et al (2010) Claudin 11 deficiency in mice results in loss of the Sertoli cell epithelial phenotype in the testis. Biol Reprod 82:202–213. https://doi.org/10.1095/biolreprod.109.078907

    Article  CAS  PubMed  Google Scholar 

  85. Gow A, Southwood CM, Li JS et al (1999) CNS Myelin and Sertoli cell tight junction strands are absent in OSP/claudin-11 null mice. Cell 99:649–659. https://doi.org/10.1016/S0092-8674(00)81553-6

    Article  CAS  PubMed  Google Scholar 

  86. Wang RS, Yeh S, Chen LM et al (2006) Androgen receptor in Sertoli cell is essential for germ cell nursery and junctional complex formation in mouse testes. Endocrinology 147:5624–5633. https://doi.org/10.1210/en.2006-0138

    Article  CAS  PubMed  Google Scholar 

  87. Willems A, Batlouni SR, Esnal A et al (2010) Selective ablation of the androgen receptor in mouse Sertoli cells affects sertoli cell maturation, barrier formation and cytoskeletal development. PLoS One 5:e14168. https://doi.org/10.1371/journal.pone.0014168

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yan HHN, Mruk DD, Lee WM, Cheng CY (2008) Blood-testis barrier dynamics are regulated by testosterone and cytokines via their differential effects on the kinetics of protein endocytosis and recycling in Sertoli cells. FASEB J 22:1945–1959. https://doi.org/10.1096/fj.06-070342

    Article  CAS  PubMed  Google Scholar 

  89. Hogarth CA, Griswold MD (2010) The key role of vitamin A in spermatogenesis. J Clin Invest 120:956–962. https://doi.org/10.1172/JCI41303

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Nicholls PK, Harrison CA, Rainczuk KE et al (2013) Retinoic acid promotes Sertoli cell differentiation and antagonises activin-induced proliferation. Mol Cell Endocrinol 377:33–43. https://doi.org/10.1016/j.mce.2013.06.034

    Article  CAS  PubMed  Google Scholar 

  91. Hasegawa K, Saga Y (2012) Retinoic acid signaling in Sertoli cells regulates organization of the blood-testis barrier through cyclical changes in gene expression. Development 139:4347–4355. https://doi.org/10.1242/dev.080119

    Article  CAS  PubMed  Google Scholar 

  92. Lie PPY, Cheng CY, Mruk DD (2013) Signalling pathways regulating the blood–testis barrier. Int J Biochem Cell Biol 45:621–625. https://doi.org/10.1016/j.biocel.2012.12.009

    Article  CAS  PubMed  Google Scholar 

  93. Lie PPY, Cheng CY, Mruk DD (2011) Interleukin-1α is a regulator of the blood-testis barrier. FASEB J 25:1244–1253. https://doi.org/10.1096/fj.10-169995

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Lui W, Lee WM, Cheng CY (2003) Transforming growth factor beta3 regulates the dynamics of Sertoli cell tight junctions via the p38 mitogen-activated protein kinase pathway. Biol Reprod 68:1597–1612. https://doi.org/10.1095/biolreprod.102.011387

    Article  CAS  PubMed  Google Scholar 

  95. Wong EWP, Mruk DD, Lee WM, Cheng CY (2010) Regulation of blood-testis barrier dynamics by TGF-3 is a Cdc42-dependent protein trafficking event. Proc Natl Acad Sci 107:11399–11404. https://doi.org/10.1073/pnas.1001077107

    Article  PubMed  PubMed Central  Google Scholar 

  96. Nicholls PK, Stanton PG, Chen JL et al (2012) Activin signaling regulates sertoli cell differentiation and function. Endocrinology 153:6065–6077. https://doi.org/10.1210/en.2012-1821

    Article  CAS  PubMed  Google Scholar 

  97. Li SYT, Yan M, Chen H et al (2018) mTORC1/rpS6 regulates blood-testis barrier dynamics and spermatogenetic function in the testis in vivo. Am J Physiol Metab 314:E174–E190. https://doi.org/10.1152/ajpendo.00263.2017

    Article  CAS  Google Scholar 

  98. De Rooij DG (2009) The spermatogonial stem cell niche. Microsc Res Tech 72:580–585

    Article  PubMed  Google Scholar 

  99. Chen S-R, Liu Y-X (2015) Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. Reproduction 149:R159–R167. https://doi.org/10.1530/REP-14-0481

    Article  CAS  PubMed  Google Scholar 

  100. Oatley JM, Oatley MJ, Avarbock MR et al (2009) Colony stimulating factor 1 is an extrinsic stimulator of mouse spermatogonial stem cell self-renewal. Development 136:1191–1199. https://doi.org/10.1242/dev.032243

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Meng X, Lindahl M, Hyvönen ME et al (2000) Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science (80–) 287:1489–1493. https://doi.org/10.1126/science.287.5457.1489

  102. Kubota H, Avarbock MR, Brinster RL (2004) Growth factors essential for self-renewal and expansion of mouse spermatogonial stem cells. Proc Natl Acad Sci 101:16489–16494. https://doi.org/10.1073/pnas.0407063101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Hofmann MC, Braydich-Stolle L, Dym M (2005) Isolation of male germ-line stem cells; Influence of GDNF. Dev Biol 279:114–124. https://doi.org/10.1016/j.ydbio.2004.12.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Naughton CK, Jain S, Strickland AM et al (2006) Glial cell-line derived neurotrophic factor-mediated RET signaling regulates spermatogonial stem cell fate. Biol Reprod 74:314–321. https://doi.org/10.1095/biolreprod.105.047365

    Article  CAS  PubMed  Google Scholar 

  105. He Z, Jiang J, Hofmann M-C, Dym M (2007) Gfra1 silencing in mouse spermatogonial stem cells results in their differentiation via the inactivation of RET tyrosine kinase. Biol Reprod 77:723–733. https://doi.org/10.1095/biolreprod.107.062513

    Article  CAS  PubMed  Google Scholar 

  106. Oatley JM, Avarbock MR, Telaranta AI et al (2006) Identifying genes important for spermatogonial stem cell self-renewal and survival. Proc Natl Acad Sci 103:9524–9529. https://doi.org/10.1073/pnas.0603332103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Masaki K, Sakai M, Kuroki S et al (2018) FGF2 has distinct molecular functions from GDNF in the mouse germline niche. Stem Cell Rep 10:1782–1792. https://doi.org/10.1016/j.stemcr.2018.03.016

    Article  CAS  Google Scholar 

  108. Takashima S, Kanatsu-Shinohara M, Tanaka T et al (2015) Functional differences between GDNF-dependent and FGF2-dependent mouse spermatogonial stem cell self-renewal. Stem Cell Reports 4:489–502. https://doi.org/10.1016/j.stemcr.2015.01.010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Heckmann L, Pock T, Tröndle I, Neuhaus N (2018) The C-X-C signalling system in the rodent vs primate testis: impact on germ cell niche interaction. Reproduction 155:R211–R219. https://doi.org/10.1530/REP-17-0617

    Article  CAS  PubMed  Google Scholar 

  110. Yang Q-E, Kim D, Kaucher A et al (2013) CXCL12-CXCR4 signaling is required for the maintenance of mouse spermatogonial stem cells. J Cell Sci 126:1009–1020. https://doi.org/10.1242/jcs.119826

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. McIver SC, Loveland KL, Roman SD et al (2013) The chemokine CXCL12 and its receptor CXCR4 are implicated in human seminoma metastasis. Andrology 1:517–529. https://doi.org/10.1111/j.2047-2927.2013.00081.x

    Article  CAS  PubMed  Google Scholar 

  112. He Z, Kokkinaki M, Dym M (2009) Signaling molecules and pathways regulating the fate of spermatogonial stem cells. Microsc Res Tech 72:586–595. https://doi.org/10.1002/jemt.20698

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Manku G, Culty M (2015) Mammalian gonocyte and spermatogonia differentiation: recent advances and remaining challenges. Reproduction 149:R139–R157. https://doi.org/10.1530/REP-14-0431

    Article  CAS  PubMed  Google Scholar 

  114. Eto K, Shiotsuki M, Sakai T, Abe S (2012) Nociceptin is upregulated by FSH signaling in Sertoli cells in murine testes. Biochem Biophys Res Commun 421:678–683. https://doi.org/10.1016/j.bbrc.2012.04.061

    Article  CAS  PubMed  Google Scholar 

  115. Lee J (2003) Temporally and spatially selective loss of Rec8 protein from meiotic chromosomes during mammalian meiosis. J Cell Sci 116:2781–2790. https://doi.org/10.1242/jcs.00495

    Article  CAS  PubMed  Google Scholar 

  116. Eto K, Shiotsuki M, Abe S (2013) Nociceptin induces Rec8 phosphorylation and meiosis in postnatal murine testes. Endocrinology 154:2891–2899. https://doi.org/10.1210/en.2012-1977

    Article  CAS  PubMed  Google Scholar 

  117. Griswold MD (2015) The initiation of spermatogenesis and the cycle of the seminiferous epithelium. In: Sertoli cell biology. Elsevier, pp 233–245

    Google Scholar 

  118. Mark M, Jacobs H, Oulad-Abdelghani M et al (2008) STRA8-deficient spermatocytes initiate, but fail to complete, meiosis and undergo premature chromosome condensation. J Cell Sci 121:3233–3242. https://doi.org/10.1242/jcs.035071

    Article  CAS  PubMed  Google Scholar 

  119. Zhang J, Eto K, Honmyou A et al (2011) Neuregulins are essential for spermatogonial proliferation and meiotic initiation in neonatal mouse testis. Development 138:3159–3168. https://doi.org/10.1242/dev.062380

    Article  CAS  PubMed  Google Scholar 

  120. Oliveira PF, Alves MG (2015) Spermatogenesis. In: Sertoli cell metabolism and spermatogenesis. Springer International Publishing, Cham, pp 15–24

    Google Scholar 

  121. Hogarth C (2015) Retinoic acid metabolism, signaling, and function in the adult testis. In: Sertoli cell biology. Elsevier, pp 247–272

    Google Scholar 

  122. O’Donnell L, Nicholls PK, O’Bryan MK et al (2011) Spermiation. Spermatogenesis 1:14–35. https://doi.org/10.4161/spmg.1.1.14525

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

This work was supported by FEDER Funds through Competitiveness and Internationalization Operational Program—COMPETE 2020 and by National Funds through FCT—Foundation for Science and Technology under the project PTDB/BBB-BQB/3804/2014. We are thankful to Institute for Biomedicine—iBiMED (UIDB/04501/2020 and POCI-01-0145-FEDER-007628) for supporting this project. iBiMED is supported by the Portuguese Foundation for Science and Technology (FCT), Compete2020 and FEDER fund. This work was also support by individual grant from FCT of the Portuguese Ministry of Science and Higher Education to JS (SFRH/BD/136896/2018), DP (SFRH/BD/137487/2018) and JVS (SFRH/BPD/123155/2016).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joana Vieira Silva .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Santiago, J., Patrício, D., Silva, J.V. (2020). Testicular Signaling: Team Work in Sperm Production. In: Silva, J.V., Freitas, M.J., Fardilha, M. (eds) Tissue-Specific Cell Signaling. Springer, Cham. https://doi.org/10.1007/978-3-030-44436-5_8

Download citation

Publish with us

Policies and ethics