#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Six Homeoproteins Directly Activate Expression in the Gene Regulatory Networks That Control Early Myogenesis


In mammals, several genetic pathways have been characterized that govern engagement of multipotent embryonic progenitors into the myogenic program through the control of the key myogenic regulatory gene Myod. Here we demonstrate the involvement of Six homeoproteins. We first targeted into a Pax3 allele a sequence encoding a negative form of Six4 that binds DNA but cannot interact with essential Eya co-factors. The resulting embryos present hypoplasic skeletal muscles and impaired Myod activation in the trunk in the absence of Myf5/Mrf4. At the axial level, we further show that Myod is still expressed in compound Six1/Six4:Pax3 but not in Six1/Six4:Myf5 triple mutant embryos, demonstrating that Six1/4 participates in the Pax3-Myod genetic pathway. Myod expression and head myogenesis is preserved in Six1/Six4:Myf5 triple mutant embryos, illustrating that upstream regulators of Myod in different embryonic territories are distinct. We show that Myod regulatory regions are directly controlled by Six proteins and that, in the absence of Six1 and Six4, Six2 can compensate.


Vyšlo v časopise: Six Homeoproteins Directly Activate Expression in the Gene Regulatory Networks That Control Early Myogenesis. PLoS Genet 9(4): e32767. doi:10.1371/journal.pgen.1003425
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003425

Souhrn

In mammals, several genetic pathways have been characterized that govern engagement of multipotent embryonic progenitors into the myogenic program through the control of the key myogenic regulatory gene Myod. Here we demonstrate the involvement of Six homeoproteins. We first targeted into a Pax3 allele a sequence encoding a negative form of Six4 that binds DNA but cannot interact with essential Eya co-factors. The resulting embryos present hypoplasic skeletal muscles and impaired Myod activation in the trunk in the absence of Myf5/Mrf4. At the axial level, we further show that Myod is still expressed in compound Six1/Six4:Pax3 but not in Six1/Six4:Myf5 triple mutant embryos, demonstrating that Six1/4 participates in the Pax3-Myod genetic pathway. Myod expression and head myogenesis is preserved in Six1/Six4:Myf5 triple mutant embryos, illustrating that upstream regulators of Myod in different embryonic territories are distinct. We show that Myod regulatory regions are directly controlled by Six proteins and that, in the absence of Six1 and Six4, Six2 can compensate.


Zdroje

1. GehringWJ, IkeoK (1999) Pax 6: mastering eye morphogenesis and eye evolution. Trends in genetics 15: 371–377.

2. KumarJP (2010) Retinal determination the beginning of eye development. Current topics in developmental biology 93: 1–28.

3. van HeyningenV, WilliamsonKA (2002) PAX6 in sensory development. Human molecular genetics 11: 1161–1167.

4. JemcJ, RebayI (2007) The eyes absent family of phosphotyrosine phosphatases: properties and roles in developmental regulation of transcription. Annual review of biochemistry 76: 513–538.

5. BuckinghamM, RelaixF (2007) The role of Pax genes in the development of tissues and organs: Pax3 and Pax7 regulate muscle progenitor cell functions. Annual review of cell and developmental biology 23: 645–673.

6. Xu PX (2012) The EYA-SO/SIX complex in development and disease. Pediatric nephrology (Berlin, Germany).

7. DuanH, ZhangC, ChenJ, SinkH, FreiE, et al. (2007) A key role of Pox meso in somatic myogenesis of Drosophila. Development (Cambridge, England) 134: 3985–3997.

8. KirbyRJ, HamiltonGM, FinneganDJ, JohnsonKJ, JarmanAP (2001) Drosophila homolog of the myotonic dystrophy-associated gene, SIX5, is required for muscle and gonad development. Current biology 11: 1044–1049.

9. LiuYH, JakobsenJS, ValentinG, AmarantosI, GilmourDT, et al. (2009) A systematic analysis of Tinman function reveals Eya and JAK-STAT signaling as essential regulators of muscle development. Developmental cell 16: 280–291.

10. LaghaM, BrunelliS, MessinaG, CumanoA, KumeT, et al. (2009) Pax3:Foxc2 reciprocal repression in the somite modulates muscular versus vascular cell fate choice in multipotent progenitors. Developmental cell 17: 892–899.

11. HeanueTA, ReshefR, DavisRJ, MardonG, OliverG, et al. (1999) Synergistic regulation of vertebrate muscle development by Dach2, Eya2, and Six1, homologs of genes required for Drosophila eye formation. Genes & development 13: 3231–3243.

12. RidgewayAG, SkerjancIS (2001) Pax3 is essential for skeletal myogenesis and the expression of Six1 and Eya2. The Journal of biological chemistry 276: 19033–19039.

13. GrifoneR, DemignonJ, HoubronC, SouilE, NiroC, et al. (2005) Six1 and Six4 homeoproteins are required for Pax3 and MRF expression during myogenesis in the mouse embryo. Development 132: 2235–2249.

14. BrownCB, EnglekaKA, WenningJ, Min LuM, EpsteinJA (2005) Identification of a hypaxial somite enhancer element regulating Pax3 expression in migrating myoblasts and characterization of hypaxial muscle Cre transgenic mice. Genesis (New York, NY) 41: 202–209.

15. GrifoneR, DemignonJ, GiordaniJ, NiroC, SouilE, et al. (2007) Eya1 and Eya2 proteins are required for hypaxial somitic myogenesis in the mouse embryo. Dev Biol 302: 602–616.

16. LinCY, ChenWT, LeeHC, YangPH, YangHJ, et al. (2009) The transcription factor Six1a plays an essential role in the craniofacial myogenesis of zebrafish. Developmental biology 331: 152–166.

17. GuoC, SunY, ZhouB, AdamRM, LiX, et al. (2011) A Tbx1-Six1/Eya1-Fgf8 genetic pathway controls mammalian cardiovascular and craniofacial morphogenesis. The Journal of clinical investigation 121: 1585–1595.

18. SambasivanR, KurataniS, TajbakhshS (2011) An eye on the head: the development and evolution of craniofacial muscles. Development (Cambridge, England) 138: 2401–2415.

19. BuckinghamM, VincentSD (2009) Distinct and dynamic myogenic populations in the vertebrate embryo. Current opinion in genetics & development 19: 444–453.

20. TajbakhshS, RocancourtD, CossuG, BuckinghamM (1997) Redefining the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Myf-5 act upstream of MyoD. Cell 89: 127–138.

21. SatoT, RocancourtD, MarquesL, ThorsteinsdottirS, BuckinghamM (2010) A Pax3/Dmrt2/Myf5 regulatory cascade functions at the onset of myogenesis. PLoS Genet 6: e1000897 doi:10.1371/journal.pgen.1000897.

22. BorelloU, BerarducciB, MurphyP, BajardL, BuffaV, et al. (2006) The Wnt/beta-catenin pathway regulates Gli-mediated Myf5 expression during somitogenesis. Development (Cambridge, England) 133: 3723–3732.

23. BajardL, RelaixF, LaghaM, RocancourtD, DaubasP, et al. (2006) A novel genetic hierarchy functions during hypaxial myogenesis: Pax3 directly activates Myf5 in muscle progenitor cells in the limb. Genes Dev 20: 2450–2464.

24. GiordaniJ, BajardL, DemignonJ, DaubasP, BuckinghamM, et al. (2007) Six proteins regulate the activation of Myf5 expression in embryonic mouse limbs. Proc Natl Acad Sci U S A 104: 11310–11315.

25. LiuY, ChuA, ChakrounI, IslamU, BlaisA (2010) Cooperation between myogenic regulatory factors and SIX family transcription factors is important for myoblast differentiation. Nucleic acids research 38: 6857–6871.

26. SpitzF, DemignonJ, PorteuA, KahnA, ConcordetJP, et al. (1998) Expression of myogenin during embryogenesis is controlled by Six/sine oculis homeoproteins through a conserved MEF3 binding site. Proc Natl Acad Sci U S A 95: 14220–14225.

27. NiroC, DemignonJ, VincentS, LiuY, GiordaniJ, et al. (2010) Six1 and Six4 gene expression is necessary to activate the fast-type muscle gene program in the mouse primary myotome. Developmental biology 338: 168–182.

28. RichardAF, DemignonJ, SakakibaraI, PujolJ, FavierM, et al. (2011) Genesis of muscle fiber-type diversity during mouse embryogenesis relies on Six1 and Six4 gene expression. Developmental biology 359: 303–320.

29. PignoniF, HuB, ZavitzKH, XiaoJ, GarrityPA, et al. (1997) The eye-specification proteins So and Eya form a complex and regulate multiple steps in Drosophila eye development. Cell 91: 881–891.

30. OhtoH, KamadaS, TagoK, TominagaSI, OzakiH, et al. (1999) Cooperation of six and eya in activation of their target genes through nuclear translocation of Eya. Molecular and cellular biology 19: 6815–6824.

31. KawakamiK, OhtoH, IkedaK, RoederRG (1996) Structure, function and expression of a murine homeobox protein AREC3, a homologue of Drosophila sine oculis gene product, and implication in development. Nucleic acids research 24: 303–310.

32. RelaixF, PolimeniM, RocancourtD, PonzettoC, SchaferBW, et al. (2003) The transcriptional activator PAX3-FKHR rescues the defects of Pax3 mutant mice but induces a myogenic gain-of-function phenotype with ligand-independent activation of Met signaling in vivo. Genes & development 17: 2950–2965.

33. TajbakhshS, BoberE, BabinetC, PourninS, ArnoldH, et al. (1996) Gene targeting the myf-5 locus with nlacZ reveals expression of this myogenic factor in mature skeletal muscle fibres as well as early embryonic muscle. Developmental dynamics 206: 291–300.

34. SassoonD, LyonsG, WrightWE, LinV, LassarA, et al. (1989) Expression of two myogenic regulatory factors myogenin and MyoD1 during mouse embryogenesis. Nature 341: 303–307.

35. AsakuraA, LyonsGE, TapscottSJ (1995) The regulation of MyoD gene expression: conserved elements mediate expression in embryonic axial muscle. Developmental biology 171: 386–398.

36. GoldhamerDJ, BrunkBP, FaermanA, KingA, ShaniM, et al. (1995) Embryonic activation of the myoD gene is regulated by a highly conserved distal control element. Development (Cambridge, England) 121: 637–649.

37. KucharczukKL, LoveCM, DoughertyNM, GoldhamerDJ (1999) Fine-scale transgenic mapping of the MyoD core enhancer: MyoD is regulated by distinct but overlapping mechanisms in myotomal and non-myotomal muscle lineages. Development (Cambridge, England) 126: 1957–1965.

38. ChenJC, RamachandranR, GoldhamerDJ (2002) Essential and redundant functions of the MyoD distal regulatory region revealed by targeted mutagenesis. Developmental biology 245: 213–223.

39. ChenJC, GoldhamerDJ (2004) The core enhancer is essential for proper timing of MyoD activation in limb buds and branchial arches. Developmental biology 265: 502–512.

40. KawakamiK, SatoS, OzakiH, IkedaK (2000) Six family genes–structure and function as transcription factors and their roles in development. BioEssays 22: 616–626.

41. OliverG, WehrR, JenkinsNA, CopelandNG, CheyetteBN, et al. (1995) Homeobox genes and connective tissue patterning. Development (Cambridge, England) 121: 693–705.

42. KlesertTR, ChoDH, ClarkJI, MaylieJ, AdelmanJ, et al. (2000) Mice deficient in Six5 develop cataracts: implications for myotonic dystrophy. Nature genetics 25: 105–109.

43. YajimaH, MotohashiN, OnoY, SatoS, IkedaK, et al. (2010) Six family genes control the proliferation and differentiation of muscle satellite cells. Experimental cell research 316: 2932–2944.

44. Le GrandF, GrifoneR, MourikisP, HoubronC, GigaudC, et al. (2012) Six1 regulates stem cell repair potential and self-renewal during skeletal muscle regeneration. J Cell Biol 198: 815–832.

45. Kassar-DuchossoyL, Gayraud-MorelB, GomesD, RocancourtD, BuckinghamM, et al. (2004) Mrf4 determines skeletal muscle identity in Myf5:Myod double-mutant mice. Nature 431: 466–471.

46. XuPX, WooI, HerH, BeierDR, MaasRL (1997) Mouse Eya homologues of the Drosophila eyes absent gene require Pax6 for expression in lens and nasal placode. Development (Cambridge, England) 124: 219–231.

47. L'HonoreA, OuimetteJF, Lavertu-JolinM, DrouinJ (2010) Pitx2 defines alternate pathways acting through MyoD during limb and somitic myogenesis. Development (Cambridge, England) 137: 3847–3856.

48. HuP, GelesKG, PaikJH, DePinhoRA, TjianR (2008) Codependent activators direct myoblast-specific MyoD transcription. Developmental cell 15: 534–546.

49. CaoL, YuY, BilkeS, WalkerRL, MayeenuddinLH, et al. (2010) Genome-wide identification of PAX3-FKHR binding sites in rhabdomyosarcoma reveals candidate target genes important for development and cancer. Cancer research 70: 6497–6508.

50. CaoY, YaoZ, SarkarD, LawrenceM, SanchezGJ, et al. (2010) Genome-wide MyoD binding in skeletal muscle cells: a potential for broad cellular reprogramming. Developmental cell 18: 662–674.

51. KressC, Vandormael-PourninS, BaldacciP, Cohen-TannoudjiM, BabinetC (1998) Nonpermissiveness for mouse embryonic stem (ES) cell derivation circumvented by a single backcross to 129/Sv strain: establishment of ES cell lines bearing the Omd conditional lethal mutation. Mammalian genome 9: 998–1001.

52. LallemandY, LuriaV, Haffner-KrauszR, LonaiP (1998) Maternally expressed PGK-Cre transgene as a tool for early and uniform activation of the Cre site-specific recombinase. Transgenic research 7: 105–112.

53. LaclefC, HamardG, DemignonJ, SouilE, HoubronC, et al. (2003) Altered myogenesis in Six1-deficient mice. Development 130: 2239–2252.

54. GrifoneR, LaclefC, SpitzF, LopezS, DemignonJ, et al. (2004) Six1 and Eya1 expression can reprogram adult muscle from the slow-twitch phenotype into the fast-twitch phenotype. Mol Cell Biol 24: 6253–6267.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 4
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#