#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

RFX2 Is a Major Transcriptional Regulator of Spermiogenesis


Failure of spermatogenesis, which is presumed to often result from genetic defects, is a common cause of male sterility. Although numerous genes associated with defects in male spermatogenesis have been identified, numerous cases of genetic male infertility remain unelucidated. We report here that the transcription factor RFX2 is a master regulator of gene expression programs required for progression through the haploid phase of spermatogenesis. Male RFX2-deficient mice are completely sterile. Spermatogenesis progresses through meiosis, but haploid cells undergo a complete block in development just prior to spermatid elongation. Gene expression profiling and ChIP-Seq analysis revealed that RFX2 controls key pathways implicated in cilium/flagellum formation, as well as genes implicated in microtubule and vesicle associated transport. The set of genes activated by RFX2 in spermatids exhibits virtually no overlap with those controlled by other known transcriptional regulators of spermiogenesis, establishing RFX2 as an essential new player in this developmental process. RFX2-deficient mice should therefore represent a valuable new model for deciphering the regulatory networks that direct sperm formation, and thereby contribute to the identification of causes of human male infertility.


Vyšlo v časopise: RFX2 Is a Major Transcriptional Regulator of Spermiogenesis. PLoS Genet 11(7): e32767. doi:10.1371/journal.pgen.1005368
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005368

Souhrn

Failure of spermatogenesis, which is presumed to often result from genetic defects, is a common cause of male sterility. Although numerous genes associated with defects in male spermatogenesis have been identified, numerous cases of genetic male infertility remain unelucidated. We report here that the transcription factor RFX2 is a master regulator of gene expression programs required for progression through the haploid phase of spermatogenesis. Male RFX2-deficient mice are completely sterile. Spermatogenesis progresses through meiosis, but haploid cells undergo a complete block in development just prior to spermatid elongation. Gene expression profiling and ChIP-Seq analysis revealed that RFX2 controls key pathways implicated in cilium/flagellum formation, as well as genes implicated in microtubule and vesicle associated transport. The set of genes activated by RFX2 in spermatids exhibits virtually no overlap with those controlled by other known transcriptional regulators of spermiogenesis, establishing RFX2 as an essential new player in this developmental process. RFX2-deficient mice should therefore represent a valuable new model for deciphering the regulatory networks that direct sperm formation, and thereby contribute to the identification of causes of human male infertility.


Zdroje

1. Matzuk MM, Lamb DJ (2008) The biology of infertility: research advances and clinical challenges. Nat Med 14: 1197–1213. doi: 10.1038/nm.f.1895 18989307

2. Visser L, Repping S (2010) Unravelling the genetics of spermatogenic failure. Reproduction 139: 303–307. doi: 10.1530/REP-09-0229 19776097

3. O'Donnell L, Meachem SJ, Stanton PG, McLaughlan RI (2005) Endocrine Regulation of Spermatogenesis. In: de Kretser DM, editor. Knobil and Neill's Physiology of Reproduction. 3rd ed. New York: Academic Press. pp. 1017–1070.

4. Bettegowda A, Wilkinson MF (2010) Transcription and post-transcriptional regulation of spermatogenesis. Philos Trans R Soc Lond, B, Biol Sci 365: 1637–1651. doi: 10.1098/rstb.2009.0196 20403875

5. Kerr JB, Loveland KL, O'Bryan MK, de Kretser DM (2005) Cytology of the Testis and Intrinsic Control Mechanisms. In: de Kretser DM, editor. Knobil and Neill's Physiology of Reproduction. 3rd ed. New York: Academic Press. pp. 827–948.

6. Dorn A, Durand B, Marfing C, Le Meur M, Benoist C, et al. (1987) Conserved major histocompatibility complex class II boxes—X and Y—are transcriptional control elements and specifically bind nuclear proteins. Proc Natl Acad Sci U S A 84: 6249–6253. 3114745

7. Aftab S, Semenec L, Chu JS, Chen N (2008) Identification and characterization of novel human tissue-specific RFX transcription factors. BMC Evol Biol 8: 226. doi: 10.1186/1471-2148-8-226 18673564

8. Emery P, Durand B, Mach B, Reith W (1996) RFX proteins, a novel family of DNA binding proteins conserved in the eukaryotic kingdom. Nucleic Acids Res 24: 803–807. 8600444

9. Gajiwala KS, Chen H, Cornille F, Roques BP, Reith W, et al. (2000) Structure of the winged-helix protein hRFX1 reveals a new mode of DNA binding. Nature 403: 916–921. 10706293

10. Swoboda P, Adler HT, Thomas JH (2000) The RFX-type transcription factor DAF-19 regulates sensory neuron cilium formation in C. elegans. Mol Cell 5: 411–421. 10882127

11. Thomas J, Morlé L, Soulavie F, Laurençon A, Sagnol S, et al. (2010) Transcriptional control of genes involved in ciliogenesis: a first step in making cilia. Biol Cell 102: 499–513. doi: 10.1042/BC20100035 20690903

12. Choksi SP, Lauter G, Swoboda P, Roy S (2014) Switching on cilia: transcriptional networks regulating ciliogenesis. Development 141: 1427–1441. doi: 10.1242/dev.074666 24644260

13. Feng C, Xu W, Zuo Z (2009) Knockout of the regulatory factor X1 gene leads to early embryonic lethality. Biochem Biophys Res Commun 386: 715–717. doi: 10.1016/j.bbrc.2009.06.111 19559676

14. Bonnafe E, Touka M, AitLounis A, Baas D, Barras E, et al. (2004) The transcription factor RFX3 directs nodal cilium development and left-right asymmetry specification. Mol Cell Biol 24: 4417–4427. 15121860

15. Benadiba C, Magnani D, Niquille M, Morlé L, Valloton D, et al. (2012) The ciliogenic transcription factor RFX3 regulates early midline distribution of guidepost neurons required for corpus callosum development. PLoS Genet 8: e1002606. doi: 10.1371/journal.pgen.1002606 22479201

16. Ait-Lounis A, Baas D, Barras E, Benadiba C, Charollais A, et al. (2007) Novel function of the ciliogenic transcription fator RFX3 in development of the endocrine pancreas. Diabetes 56: 950–959. 17229940

17. Blackshear PJ, Graves JP, Stumpo DJ, Cobos I, Rubenstein JL, et al. (2003) Graded phenotypic response to partial and complete deficiency of a brain-specific transcript variant of the winged helix transcription factor RFX4. Development 130: 4539–4552. 12925582

18. Zarbalis K, May SR, Shen Y, Ekker M, Rubenstein JL, et al. (2004) A focused and efficient genetic Screening Strategy in the mouse: Identification of mutations that disrupt cortial development. Plos Biology 2: 1177–1187.

19. Zhang D, Zeldin DC, Blackshear PJ (2007) Regulatory factor X4 variant 3: a transcription factor involved in brain development and disease. J Neurosci Res 85: 3515–3522. 17510980

20. Steimle V, Durand B, Barras E, Zufferey M, Hadam MR, et al. (1995) A novel DNA-binding regulatory factor is mutated in primary MHC class II deficiency (bare lymphocyte syndrome). Genes Dev 9: 1021–1032. 7744245

21. Smith SB, Qu H-Q, Taleb N, Kishimoto NY, Scheel DW, et al. (2010) Rfx6 directs islet formation and insulin production in mice and humans. Nature 463: 775–780. doi: 10.1038/nature08748 20148032

22. Soyer J, Flasse L, Raffelsberger W, Beucher A, Orvain C, et al. (2010) Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development. Development 137: 203–212. doi: 10.1242/dev.041673 20040487

23. Manojlovic Z, Earwood R, Kato A, Stefanovic B, Kato Y (2014) RFX7 is required for the formation of cilia in the neural tube. Mech Dev 132: 28–37. doi: 10.1016/j.mod.2014.02.001 24530844

24. Bisgrove BW, Makova S, Yost HJ, Brueckner M (2012) RFX2 is essential in the ciliated organ of asymmetry and an RFX2 transgene identifies a population of ciliated cells sufficient for fluid flow. Dev Biol 363: 166–178. doi: 10.1016/j.ydbio.2011.12.030 22233545

25. Chung M-I, Kwon T, Tu F, Brooks ER, Gupta R, et al. (2014) Coordinated genomic control of ciliogenesis and cell movement by RFX2. Elife 3: e01439. doi: 10.7554/eLife.01439 24424412

26. Chung M-I, Peyrot SM, Leboeuf S, Park TJ, Mcgary KL, et al. (2012) RFX2 is broadly required for ciliogenesis during vertebrate development. Dev Biol 363: 155–165. doi: 10.1016/j.ydbio.2011.12.029 22227339

27. Liu Y, Pathak N, Kramer-Zucker A, Drummond IA (2007) Notch signaling controls the differentiation of transporting epithelia and multiciliated cells in the zebrafish pronephros. Development 134: 1111–1122. 17287248

28. Reith W, Ucla C, Barras E, Gaud A, Durand B, et al. (1994) RFX1, a transactivator of hepatitis B virus enhancer I, belongs to a novel family of homodimeric and heterodimeric DNA-binding proteins. Mol Cell Biol 14: 1230–1244. 8289803

29. Laiho A, Kotaja N, Gyenesei A, Sironen A (2013) Transcriptome profiling of the murine testis during the first wave of spermatogenesis. PLoS ONE 8: e61558. doi: 10.1371/journal.pone.0061558 23613874

30. Schultz N, Hamra FK, Garbers DL (2003) A multitude of genes expressed solely in meiotic or spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci U S A 100: 12201–12206. 14526100

31. Shima JE, McLean DJ, McCarrey JR, Griswold MD (2004) The murine testicular transcriptome: characterizing gene expression in the testis during the progression of spermatogenesis. Biol Reprod 71: 319–330. 15028632

32. Horvath GC, Kistler WS, Kistler MK (2004) RFX2 is a potential transcriptional regulatory factor for histone H1t and other genes expressed during the meiotic phase of spermatogenesis. Biol Reprod 71: 1551–1559. 15229132

33. Horvath GC, Kistler MK, Kistler WS (2009) RFX2 is a candidate downstream amplifier of A-MYB regulation in mouse spermatogenesis. BMC Dev Biol 9: 63. doi: 10.1186/1471-213X-9-63 20003220

34. Kim MJ, Li D, Cui Y, Mueller K, Chears WC, et al. (2006) Regulatory factor interactions and somatic silencing of the germ cell-specific ALF gene. J Biol Chem 281: 34288–34298. 16966320

35. Toscani A, Mettus RV, Coupland R, Simpkins H, Litvin J, et al. (1997) Arrest of spermatogenesis and defective breast development in mice lacking A-myb. Nature 386: 713–717. 9109487

36. Bolcun-Filas E, Bannister LA, Barash A, Schimenti KJ, Hartford SA, et al. (2011) A-MYB (MYBL1) transcription factor is a master regulator of male meiosis. Development 138: 3319–3330. doi: 10.1242/dev.067645 21750041

37. Kistler WS, Horvath GC, Dasgupta A, Kistler MK (2009) Differential expression of Rfx1-4 during mouse spermatogenesis. Gene Expr Patterns 9: 515–519. doi: 10.1016/j.gep.2009.07.004 19596083

38. Hogeveen KN, Sassone-Corsi P (2006) Regulation of gene expression in post-meiotic male germ cells: CREM-signalling pathways and male fertility. Hum Fertil (Camb) 9: 73–79.

39. Kosir R, Juvan P, Perse M, Budefeld T, Majdic G, et al. (2012) Novel Insights into the Downstream Pathways and Targets Controlled by Transcription Factors CREM in the Testis. PLoS ONE 7: e31798. doi: 10.1371/journal.pone.0031798 22384077

40. Zhou H, Grubisic I, Zheng K, He Y, Wang PJ, et al. (2013) Taf7l cooperates with Trf2 to regulate spermiogenesis. Proc Natl Acad Sci USA 110: 16886–16891. doi: 10.1073/pnas.1317034110 24082143

41. MacGregor GR, Russell LD, Van Beek MEAB, Hanten GR, Kovac MJ, et al. (1990) Symplastic spermatids (sys): A recessive insertional mutation in mice causing a defect in spermatogenesis. Proc Natl Acad Sci USA 87: 5016–5020. 2164218

42. Meistrich ML, Mohapatra B, Shirley CR, Zhao M (2003) Roles of transition nuclear proteins in spermiogenesis. Chromosoma 111: 483–488. 12743712

43. Clermont Y, Oko R, Hermo L (1993) Cell Biology of Mammalian Spermatogenesis. In: Desjardins C, Ewing LL, editors. Cell and Molecular Biology of the Testis. New York: Oxford University Press. pp. 332–376.

44. Russell LD, Ettlin RA, Sinha Hikim AP, Clegg ED (1990) Histological and Histopathological Evaluation of the Testis. Clearwater: Cache River Press.

45. Berruti G, Paiardi C (2011) Acrosome biogenesis: Revisiting old questions to yield new insights. Spermatogenesis 1: 95–98. 22319656

46. Kierszenbaum AL, Tres LL, Rivkin E, Kang-Decker N, van Deursen JMA (2004) The acroplaxome is the docking site of Golgi-derived myosin Va/Rab27a/b- containing proacrosomal vesicles in wild-type and Hrb mutant mouse spermatids. Biol Reprod 70: 1400–1410. 14724135

47. Moreno RD, Palomino J, Schatten G (2006) Assembly of spermatid acrosome depends on microtubule organization during mammalian spermiogenesis. Dev Biol 293: 218–227. 16540102

48. Sperry AO (2012) The dynamic cytoskeleton of the developing male germ cell. Biol Cell 104: 297–305. doi: 10.1111/boc.201100102 22276751

49. Lee NP, Cheng CY (2004) Ectoplasmic specialization, a testis-specific actin-based adherens junction type: is this a potential target for male contraceptive development? Hum Reprod Update 10: 349–369. 15192055

50. Russell LD, Tallon-Doran M, Weber JE, Wong V, Peterson RN (1983) Three-dimensional reconstruction of a rat stage V Sertoli Cell: III. A study of specific cellular relationships. Am J Anat 167: 181–192. 6613903

51. Dym M, Fawcett DW (1971) Further observations on the numbers of spermatogonia, spermatocytes, and spermatids connected by intercellular bridges in the mammalian testis. Biol Reprod 4: 195–215. 4107186

52. Kierszenbaum AL, Tres LL (2004) The acrosome-acroplaxome-manchette complex and the shaping of the spermatid head. Arch Histol Cytol 67: 271–284. 15700535

53. Meistrich ML (1993) Nuclear Morphogenesis during Spermiogenesis. In: de Kretser DM, editor. Molecular Biology of the Male Reproductive System. New York: Academic Press. pp. 67–97.

54. Arnaiz O, Malinowska A, Kolotz C, L S, Dadlez M, et al. (2009) Cildb: a knowledgebase for centrosomes and cilia. Database (Oxford) 2009: bap022.

55. van Dam TJ, Wheway G, Slaats GG, Group SS, Huynen MA, et al. (2013) The SYSCILIA gold standard (SCGSv1) of known ciliary components and its applications within a systems biology consortium. Cilia 2: 7. doi: 10.1186/2046-2530-2-7 23725226

56. Obholz KL, Akopyan A, Waymire KG, MacGregor GR (2006) FNDC3A is required for adhesion between spermatids and Sertoli cells. Dev Biol 298: 498–513. 16904100

57. Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY (2014) Fascin 1 is an actin filament-bundling protein that regulates ectoplasmic specialization dynamics in the rat testis. Am J Physiol Endocrinol Metab 307: E738–753. doi: 10.1152/ajpendo.00113.2014 25159326

58. Yan W (2009) Male infertility caused by spermiogenic defects: lessons from gene knockouts. Molec Cell Endocrinol 306: 24–32. doi: 10.1016/j.mce.2009.03.003 19481682

59. Martianov I, Fimia GM, Dierich A, Parvinen M, Sassone-Corsi P, et al. (2001) Late arrest of spermiogenesis and germ cell apoptosis in mice lacking the TBP-like TLF/TRF2 gene. Mol Cell 7: 509–515. 11463376

60. Blendy JA, Kaestner KH, Weinbauer GF, Nieschlag E, Schütz G (1996) Severe impairment of spermatogenesis in mice lacking the CREM gene. Nature 380: 162–165. 8600391

61. Nantel F, Monaco L, Foulkes NS, Masquilier D, LeMeur M, et al. (1996) Spermiogenesis deficiency and germ-cell apoptosis in CREM-mutant mice. Nature 380: 159–162. 8600390

62. Nantel F, Sassone-Corsi P (1996) A transcriptional master switch during the spermatogenesis differentiation program. Front Biosci 1: d266–269. 9159233

63. Martianov I, Choukrallah MA, Krebs A, Ye T, Legras S, et al. (2010) Cell-specific occupancy of an extended repertoire of CREM and CREB binding loci in male germ cells. BMC Genomics 11: 530. doi: 10.1186/1471-2164-11-530 20920259

64. Kolthur-Seethaaram U, Martianov I, Davidson I (2008) Specialization of the general transcriptional machinery in male germ cells. Cell Cycle 7: 3493–3498. 19001848

65. Falender AE, Freiman RN, Geles KG, Lo KC, Hwang K, et al. (2005) Maintenance of spermatogenesis requires TAF4b, a gonad-specific subunit of TFIID. Genes Dev 19: 794–803. 15774719

66. Upadhyaya AB, Lee SH, DeJong J (1999) Identification of a general transcription factor TFIIAa/b homolog selectively expressed in testis. J Biol Chem 274: 18040–18048. 10364255

67. White-Cooper H, Davidson I (2011) Unique aspects of transcription regulation in male germ cells. Cold Spring Harb Perspect Biol 3: a002626. doi: 10.1101/cshperspect.a002626 21555408

68. Laurencon A, Dubruille R, Efimenko E, Grenier G, Bissett R, et al. (2007) Identification of novel regulatory factor X (RFX) target genes by comparative genomics in Drosophila species. Genome Biol 8: R195. 17875208

69. Piasecki BP, Burghoorn J, Swoboda P (2010) Regulatory Factor X (RFX)-mediated transcriptional rewiring of ciliary genes in animals. Proc Natl Acad Sci U S A 107: 12969–12974. doi: 10.1073/pnas.0914241107 20615967

70. Garcia-Gonzalo FR, Reiter JF (2012) Scoring a backstage pass: mechanisms of ciliogenesis and ciliary access. J Cell Biol 197: 697–709. doi: 10.1083/jcb.201111146 22689651

71. Reiter JF, Blacque OE, Leroux MR (2012) The base of the cilium: roles for transition fibres and the transition zone in ciliary formation, maintenance and compartmentalization. EMBO Rep 13: 608–618. doi: 10.1038/embor.2012.73 22653444

72. Lobo LJ, Zariwala MA, Noone PG (2014) Primary ciliary dyskinesia. QJM 107: 691–699. doi: 10.1093/qjmed/hcu063 24652656

73. Meikar O, Vagin VV, Chalmel F, Sõstar K, Lardenois A, et al. (2014) An atlas of chromatoid body components. RNA 20: 483–495. doi: 10.1261/rna.043729.113 24554440

74. Castañeda J, Genzor P, van der Heijden GW, Sarkeshik A, Yates JR 3rd, et al. (2014) Reduced pachytene piRNAs and translation underlie spermiogenic arrest in Maelstrom mutant mice. EMBO J.

75. Li XZ, Roy CK, Dong X, Bolcun-Filas E, Wang J, et al. (2013) An ancient transcription factor initiates the burst of piRNA production during early meiosis in mouse testes. Mol Cell 50: 67–81. doi: 10.1016/j.molcel.2013.02.016 23523368

76. Wong EW, Cheng CY (2009) Polarity proteins and cell-cell interactions in the testis. Int Rev Cell Mol Biol 278: 309–353. doi: 10.1016/S1937-6448(09)78007-4 19815182

77. Gliki G, Ebnet K, Aurrand-Lions M, Imhof BA, Adams RH (2004) Spermatid differentiation requires the assembly of a cell polarity complex downstream of junctional adhesion molecule-C. Nature 431: 320–324. 15372036

78. O'Donnell L, O'Bryan MK (2014) Microtubules and spermatogenesis. Semin Cell Dev Biol.

79. Sugioka K, Sawa H (2012) Formation and functions of asymmetric microtubule organization in polarized cells. Curr Opin Cell Biol 24: 517–525. doi: 10.1016/j.ceb.2012.05.007 22704717

80. Rodriguez CI, Buchholz F, Galloway J, Sequerra R, Kasper J, et al. (2000) High-efficiency deleter mice show that FLPe is an alternative to Cre-loxP. Nat Genet 25: 139–140. 10835623

81. Ma W, Horvath GC, Kistler MK, Kistler WS (2008) Expression patterns of SP1 and SP3 during mouse spermatogenesis: SP1 down-regulation correlates with two successive promoter changes and translationally compromised transcripts. Biol Reprod 79: 289–300. doi: 10.1095/biolreprod.107.067082 18417714

82. Oko RJ, Jando V, Wagner CL, Kistler WS, Hermo LS (1996) Chromatin reorganization in rat spermatids during the disappearance of testis-specific histone, H1t, and the appearance of transition proteins TP1 and TP2. Biol Reprod 54: 1141–1157. 8722637

83. Hann C, Behrmann I (2007) A cost effective non-commercial ECL-solution for Western blot detections yielding strong signals and low background. J Immunol Methods 318: 11–19. 17141265

84. Gage GJ, Kipke DR, Shain W (2012) Whole animal perfusion fixation for rodents. J Vis Exp.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#