#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

GLI3 resides at the intersection of hedgehog and androgen action to promote male sex differentiation


Autoři: Anbarasi Kothandapani aff001;  Samantha R. Lewis aff001;  Jessica L. Noel aff001;  Abbey Zacharski aff001;  Kyle Krellwitz aff001;  Anna Baines aff001;  Stephanie Winske aff001;  Chad M. Vezina aff001;  Elena M. Kaftanovskaya aff002;  Alexander I. Agoulnik aff002;  Emily M. Merton aff003;  Martin J. Cohn aff003;  Joan S. Jorgensen aff001
Působiště autorů: Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America aff001;  Department of Human and Molecular Genetics, Florida International University, Miami, Florida, United States of America aff002;  Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America aff003
Vyšlo v časopise: GLI3 resides at the intersection of hedgehog and androgen action to promote male sex differentiation. PLoS Genet 16(6): e32767. doi:10.1371/journal.pgen.1008810
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pgen.1008810

Souhrn

Urogenital tract abnormalities are among the most common congenital defects in humans. Male urogenital development requires Hedgehog-GLI signaling and testicular hormones, but how these pathways interact is unclear. We found that Gli3XtJ mutant mice exhibit cryptorchidism and hypospadias due to local effects of GLI3 loss and systemic effects of testicular hormone deficiency. Fetal Leydig cells, the sole source of these hormones in developing testis, were reduced in numbers in Gli3XtJ testes, and their functional identity diminished over time. Androgen supplementation partially rescued testicular descent but not hypospadias in Gli3XtJ mutants, decoupling local effects of GLI3 loss from systemic effects of androgen insufficiency. Reintroduction of GLI3 activator (GLI3A) into Gli3XtJ testes restored expression of Hedgehog pathway and steroidogenic genes. Together, our results show a novel function for the activated form of GLI3 that translates Hedgehog signals to reinforce fetal Leydig cell identity and stimulate timely INSL3 and testosterone synthesis in the developing testis. In turn, exquisite timing and concentrations of testosterone are required to work alongside local GLI3 activity to control development of a functionally integrated male urogenital tract.

Klíčová slova:

Androgens – Cell differentiation – Embryos – Genital anatomy – Hedgehog signaling – Sexual differentiation – Testes – Testosterone


Zdroje

1. Thorup J, McLachlan R, Cortes D, Nation TR, Balic A, Southwell BR, et al. What is new in cryptorchidism and hypospadias—a critical review on the testicular dysgenesis hypothesis. J Pediatr Surg. 2010;45(10):2074–86. doi: 10.1016/j.jpedsurg.2010.07.030 20920735

2. Welsh M, Suzuki H, Yamada G. The masculinization programming window. Endocr Dev. 2014;27:17–27. doi: 10.1159/000363609 25247641

3. Chul Kim S, Kyoung Kwon S, Pyo Hong Y. Trends in the incidence of cryptorchidism and hypospadias of registry-based data in Korea: a comparison between industrialized areas of petrochemical estates and a non-industrialized area. Asian J Androl. 2011;13(5):715–8. doi: 10.1038/aja.2010.53 20729869

4. Yamada G, Satoh Y, Baskin LS, Cunha GR. Cellular and molecular mechanisms of development of the external genitalia. Differentiation. 2003;71(8):445–60. doi: 10.1046/j.1432-0436.2003.7108001.x 14641326

5. Haraguchi R, Mo R, Hui C, Motoyama J, Makino S, Shiroishi T, et al. Unique functions of Sonic hedgehog signaling during external genitalia development. Development. 2001;128(21):4241–50. 11684660

6. Perriton CL, Powles N, Chiang C, Maconochie MK, Cohn MJ. Sonic hedgehog signaling from the urethral epithelium controls external genital development. Dev Biol. 2002;247(1):26–46. doi: 10.1006/dbio.2002.0668 12074550

7. Clark AM, Garland KK, Russell LD. Desert hedgehog (Dhh) gene is required in the mouse testis for formation of adult-type Leydig cells and normal development of peritubular cells and seminiferous tubules. Biol Reprod. 2000;63(6):1825–38. doi: 10.1095/biolreprod63.6.1825 11090455

8. Yao HH, Whoriskey W, Capel B. Desert Hedgehog/Patched 1 signaling specifies fetal Leydig cell fate in testis organogenesis. Genes Dev. 2002;16(11):1433–40. doi: 10.1101/gad.981202 12050120

9. Haraguchi R, Motoyama J, Sasaki H, Satoh Y, Miyagawa S, Nakagata N, et al. Molecular analysis of coordinated bladder and urogenital organ formation by Hedgehog signaling. Development. 2007;134(3):525–33. doi: 10.1242/dev.02736 17202190

10. Miyagawa S, Matsumaru D, Murashima A, Omori A, Satoh Y, Haraguchi R, et al. The role of sonic hedgehog-Gli2 pathway in the masculinization of external genitalia. Endocrinology. 2011;152(7):2894–903. doi: 10.1210/en.2011-0263 21586556

11. He F, Akbari P, Mo R, Zhang JJ, Hui CC, Kim PC, et al. Adult Gli2+/-;Gli3Delta699/+ Male and Female Mice Display a Spectrum of Genital Malformation. PLoS One. 2016;11(11):e0165958. doi: 10.1371/journal.pone.0165958 27814383

12. Barsoum I, Yao HH. Redundant and Differential Roles of Transcription Factors Gli1 and Gli2 in the Development of Mouse Fetal Leydig Cells1. Biol Reprod. 2011;84(5):894–9. doi: 10.1095/biolreprod.110.088997 21209421

13. Narumi Y, Kosho T, Tsuruta G, Shiohara M, Shimazaki E, Mori T, et al. Genital abnormalities in Pallister-Hall syndrome: Report of two patients and review of the literature. Am J Med Genet A. 2010;152a(12):3143–7. doi: 10.1002/ajmg.a.33720 21108399

14. Ito S, Kitazawa R, Haraguchi R, Kondo T, Ouchi A, Ueda Y, et al. Novel GLI3 variant causing overlapped Greig cephalopolysyndactyly syndrome (GCPS) and Pallister-Hall syndrome (PHS) phenotype with agenesis of gallbladder and pancreas. Diagn Pathol. 2018;13(1):1. doi: 10.1186/s13000-017-0682-8 29368652

15. Hui CC, Joyner AL. A mouse model of greig cephalopolysyndactyly syndrome: the extra-toesJ mutation contains an intragenic deletion of the Gli3 gene. Nat Genet. 1993;3(3):241–6. doi: 10.1038/ng0393-241 8387379

16. Zheng Z, Cohn MJ. Developmental basis of sexually dimorphic digit ratios. Proc Natl Acad Sci U S A. 2011;108(39):16289–94. doi: 10.1073/pnas.1108312108 21896736

17. Bushman W. Hedgehog Signaling in Prostate Development, Regeneration and Cancer. J Dev Biol. 2016;4(4). doi: 10.3390/jdb4040030 29615593

18. Sadeghian H, Anand-Ivell R, Balvers M, Relan V, Ivell R. Constitutive regulation of the Insl3 gene in rat Leydig cells. Mol Cell Endocrinol. 2005;241(1–2):10–20. doi: 10.1016/j.mce.2005.03.017 16006031

19. Suzuki H, Suzuki K, Yamada G. Systematic analyses of murine masculinization processes based on genital sex differentiation parameters. Dev Growth Differ. 2015;57(9):639–47. doi: 10.1111/dgd.12247 26660623

20. Haraguchi R, Suzuki K, Murakami R, Sakai M, Kamikawa M, Kengaku M, et al. Molecular analysis of external genitalia formation: the role of fibroblast growth factor (Fgf) genes during genital tubercle formation. Development. 2000;127(11):2471–9. 10804187

21. Zheng Z, Armfield BA, Cohn MJ. Timing of androgen receptor disruption and estrogen exposure underlies a spectrum of congenital penile anomalies. Proc Natl Acad Sci U S A. 2015;112(52):E7194–203. doi: 10.1073/pnas.1515981112 26598695

22. Hutson JM, Hasthorpe S, Heyns CF. Anatomical and functional aspects of testicular descent and cryptorchidism. Endocr Rev. 1997;18(2):259–80. doi: 10.1210/edrv.18.2.0298 9101140

23. Emmen JM, McLuskey A, Grootegoed JA, Brinkmann AO. Androgen action during male sex differentiation includes suppression of cranial suspensory ligament development. Hum Reprod. 1998;13(5):1272–80. doi: 10.1093/humrep/13.5.1272 9647559

24. Hughes IA, Acerini CL. Factors controlling testis descent. Eur J Endocrinol. 2008;159 Suppl 1:S75–82.

25. Franco HL, Yao HH. Sex and hedgehog: roles of genes in the hedgehog signaling pathway in mammalian sexual differentiation. Chromosome Res. 2012;20(1):247–58. doi: 10.1007/s10577-011-9254-z 22105695

26. Adham IM, Emmen JM, Engel W. The role of the testicular factor INSL3 in establishing the gonadal position. Mol Cell Endocrinol. 2000;160(1–2):11–6. doi: 10.1016/s0303-7207(99)00188-4 10715534

27. Emmen JM, McLuskey A, Adham IM, Engel W, Grootegoed JA, Brinkmann AO. Hormonal control of gubernaculum development during testis descent: gubernaculum outgrowth in vitro requires both insulin-like factor and androgen. Endocrinology. 2000;141(12):4720–7. doi: 10.1210/endo.141.12.7830 11108287

28. Bogatcheva NV, Truong A, Feng S, Engel W, Adham IM, Agoulnik AI. GREAT/LGR8 is the only receptor for insulin-like 3 peptide. Mol Endocrinol. 2003;17(12):2639–46. doi: 10.1210/me.2003-0096 12933905

29. van der Schoot P, Emmen JM. Development, structure and function of the cranial suspensory ligaments of the mammalian gonads in a cross-species perspective; their possible role in effecting disturbed testicular descent. Hum Reprod Update. 1996;2(5):399–418. doi: 10.1093/humupd/2.5.399 15717439

30. Wegner KA, Keikhosravi A, Eliceiri KW, Vezina CM. Fluorescence of Picrosirius Red Multiplexed With Immunohistochemistry for the Quantitative Assessment of Collagen in Tissue Sections. J Histochem Cytochem. 2017;65(8):479–90. doi: 10.1369/0022155417718541 28692327

31. Bredfeldt JS, Liu Y, Pehlke CA, Conklin MW, Szulczewski JM, Inman DR, et al. Computational segmentation of collagen fibers from second-harmonic generation images of breast cancer. J Biomed Opt. 2014;19(1):16007. doi: 10.1117/1.JBO.19.1.016007 24407500

32. Lejeune H, Habert R, Saez JM. Origin, proliferation and differentiation of Leydig cells. J Mol Endocrinol. 1998;20(1):1–25. doi: 10.1677/jme.0.0200001 9513078

33. Kaftanovskaya EM, Lopez C, Ferguson L, Myhr C, Agoulnik AI. Genetic ablation of androgen receptor signaling in fetal Leydig cell lineage affects Leydig cell functions in adult testis. Faseb j. 2015;29(6):2327–37. doi: 10.1096/fj.14-263632 25713029

34. Wen Q, Zheng QS, Li XX, Hu ZY, Gao F, Cheng CY, et al. Wt1 dictates the fate of fetal and adult Leydig cells during development in the mouse testis. Am J Physiol Endocrinol Metab. 2014;307(12):E1131–43. doi: 10.1152/ajpendo.00425.2014 25336526

35. Chen M, Zhang L, Cui X, Lin X, Li Y, Wang Y, et al. Wt1 directs the lineage specification of sertoli and granulosa cells by repressing Sf1 expression. Development. 2017;144(1):44–53. doi: 10.1242/dev.144105 27888191

36. Qin J, Tsai MJ, Tsai SY. Essential roles of COUP-TFII in Leydig cell differentiation and male fertility. PLoS One. 2008;3(9):e3285. doi: 10.1371/journal.pone.0003285 18818749

37. Tang H, Brennan J, Karl J, Hamada Y, Raetzman L, Capel B. Notch signaling maintains Leydig progenitor cells in the mouse testis. Development. 2008;135(22):3745–53. doi: 10.1242/dev.024786 18927153

38. Brennan J, Tilmann C, Capel B. Pdgfr-alpha mediates testis cord organization and fetal Leydig cell development in the XY gonad. Genes Dev. 2003;17(6):800–10. doi: 10.1101/gad.1052503 12651897

39. Gerhardt C, Wiegering A, Leu T, Ruther U. Control of Hedgehog Signalling by the Cilia-Regulated Proteasome. J Dev Biol. 2016;4(3).

40. Choksi SP, Lauter G, Swoboda P, Roy S. Switching on cilia: transcriptional networks regulating ciliogenesis. Development. 2014;141(7):1427–41. doi: 10.1242/dev.074666 24644260

41. Park SY, Tong M, Jameson JL. Distinct roles for steroidogenic factor 1 and desert hedgehog pathways in fetal and adult Leydig cell development. Endocrinology. 2007;148(8):3704–10. doi: 10.1210/en.2006-1731 17495005

42. Baba T, Otake H, Inoue M, Sato T, Ishihara Y, Moon JY, et al. Ad4BP/SF-1 regulates cholesterol synthesis to boost the production of steroids. Commun Biol. 2018;1:18. doi: 10.1038/s42003-018-0020-z 30271905

43. Morohashi KI, Omura T. Ad4BP/SF-1, a transcription factor essential for the transcription of steroidogenic cytochrome P450 genes and for the establishment of the reproductive function. Faseb j. 1996;10(14):1569–77. doi: 10.1096/fasebj.10.14.9002548 9002548

44. Tang C, Pan Y, Luo H, Xiong W, Zhu H, Ruan H, et al. Hedgehog signaling stimulates the conversion of cholesterol to steroids. Cell Signal. 2015;27(3):487–97. doi: 10.1016/j.cellsig.2015.01.004 25582983

45. Gao L, Kim Y, Kim B, Lofgren SM, Schultz-Norton JR, Nardulli AM, et al. Two regions within the proximal steroidogenic factor 1 promoter drive somatic cell-specific activity in developing gonads of the female mouse. Biol Reprod. 2011;84(3):422–34. doi: 10.1095/biolreprod.110.084590 20962249

46. Shin SH, Kogerman P, Lindstrom E, Toftgard R, Biesecker LG. GLI3 mutations in human disorders mimic Drosophila cubitus interruptus protein functions and localization. Proc Natl Acad Sci U S A. 1999;96(6):2880–4. doi: 10.1073/pnas.96.6.2880 10077605

47. Skakkebaek NE, Rajpert-De Meyts E, Main KM. Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects. Hum Reprod. 2001;16(5):972–8. doi: 10.1093/humrep/16.5.972 11331648

48. Chen Y, Yu H, Pask AJ, Fujiyama A, Suzuki Y, Sugano S, et al. Hormone-responsive genes in the SHH and WNT/beta-catenin signaling pathways influence urethral closure and phallus growth. Biol Reprod. 2018;99(4):806–16. doi: 10.1093/biolre/ioy117 29767687

49. Miyagawa S, Satoh Y, Haraguchi R, Suzuki K, Iguchi T, Taketo MM, et al. Genetic interactions of the androgen and Wnt/beta-catenin pathways for the masculinization of external genitalia. Mol Endocrinol. 2009;23(6):871–80. doi: 10.1210/me.2008-0478 19282366

50. Murashima A, Kishigami S, Thomson A, Yamada G. Androgens and mammalian male reproductive tract development. Biochim Biophys Acta. 2015;1849(2):163–70. doi: 10.1016/j.bbagrm.2014.05.020 24875095

51. Lin C, Yin Y, Veith GM, Fisher AV, Long F, Ma L. Temporal and spatial dissection of Shh signaling in genital tubercle development. Development. 2009;136(23):3959–67. doi: 10.1242/dev.039768 19906863

52. Seifert AW, Bouldin CM, Choi KS, Harfe BD, Cohn MJ. Multiphasic and tissue-specific roles of sonic hedgehog in cloacal septation and external genitalia development. Development. 2009;136(23):3949–57. doi: 10.1242/dev.042291 19906862

53. Cunha GR, Baskin L. Development of human male and female urogenital tracts. Differentiation. 2018;103:1–4. doi: 10.1016/j.diff.2018.09.002 30262219

54. Doles J, Cook C, Shi X, Valosky J, Lipinski R, Bushman W. Functional compensation in Hedgehog signaling during mouse prostate development. Dev Biol. 2006;295(1):13–25. doi: 10.1016/j.ydbio.2005.12.002 16707121

55. Toivanen R, Shen MM. Prostate organogenesis: tissue induction, hormonal regulation and cell type specification. Development. 2017;144(8):1382–98. doi: 10.1242/dev.148270 28400434

56. Allgeier SH, Lin TM, Moore RW, Vezina CM, Abler LL, Peterson RE. Androgenic regulation of ventral epithelial bud number and pattern in mouse urogenital sinus. Dev Dyn. 2010;239(2):373–85. doi: 10.1002/dvdy.22169 19941349

57. Tremblay JJ. What signals testis descent? Biol Reprod. 2010;83(5):687–9. doi: 10.1095/biolreprod.110.087197 20668256

58. van der Schoot P, Elger W. Androgen-induced prevention of the outgrowth of cranial gonadal suspensory ligaments in fetal rats. J Androl. 1992;13(6):534–42. 1293133

59. Kassim NM, Russell DA, Payne AP. Does the cranial suspensory ligament have a role in cryptorchidism? Cells Tissues Organs. 2010;191(4):307–15. doi: 10.1159/000260062 19940435

60. Hammes A, Andreassen TK, Spoelgen R, Raila J, Hubner N, Schulz H, et al. Role of endocytosis in cellular uptake of sex steroids. Cell. 2005;122(5):751–62. doi: 10.1016/j.cell.2005.06.032 16143106

61. Yao HH, Capel B. Disruption of testis cords by cyclopamine or forskolin reveals independent cellular pathways in testis organogenesis. Dev Biol. 2002;246(2):356–65. doi: 10.1006/dbio.2002.0663 12051821

62. Shima Y, Miyabayashi K, Baba T, Otake H, Katsura Y, Oka S, et al. Identification of an enhancer in the Ad4BP/SF-1 gene specific for fetal Leydig cells. Endocrinology. 2012;153(1):417–25. doi: 10.1210/en.2011-1407 22128023

63. Buscher D, Grotewold L, Ruther U. The XtJ allele generates a Gli3 fusion transcript. Mamm Genome. 1998;9(8):676–8. doi: 10.1007/s003359900845 9680393

64. Keil KP, Mehta V, Abler LL, Joshi PS, Schmitz CT, Vezina CM. Visualization and quantification of mouse prostate development by in situ hybridization. Differentiation. 2012;84(3):232–9. doi: 10.1016/j.diff.2012.07.005 22898663

65. Rasmussen FE, Wiltbank MC, Christensen JO, Grummer RR. Effects of fenprostalene and estradiol-17 beta benzoate on parturition and retained placenta in dairy cows and heifers. J Dairy Sci. 1996;79(2):227–34. doi: 10.3168/jds.S0022-0302(96)76355-5 8708084

66. Lee J, Foong YH, Musaitif I, Tong T, Jefcoate C. Analysis of specific RNA in cultured cells through quantitative integration of q-PCR and N-SIM single cell FISH images: Application to hormonal stimulation of StAR transcription. Mol Cell Endocrinol. 2016;429:93–105. doi: 10.1016/j.mce.2016.04.001 27091298


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 6
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#