#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Oct4 Is Required ∼E7.5 for Proliferation in the Primitive Streak


Oct4 is a widely recognized pluripotency factor as it maintains Embryonic Stem (ES) cells in a pluripotent state, and, in vivo, prevents the inner cell mass (ICM) in murine embryos from differentiating into trophectoderm. However, its function in somatic tissue after this developmental stage is not well characterized. Using a tamoxifen-inducible Cre recombinase and floxed alleles of Oct4, we investigated the effect of depleting Oct4 in mouse embryos between the pre-streak and headfold stages, ∼E6.0–E8.0, when Oct4 is found in dynamic patterns throughout the embryonic compartment of the mouse egg cylinder. We found that depletion of Oct4 ∼E7.5 resulted in a severe phenotype, comprised of craniorachischisis, random heart tube orientation, failed turning, defective somitogenesis and posterior truncation. Unlike in ES cells, depletion of the pluripotency factors Sox2 and Oct4 after E7.0 does not phenocopy, suggesting that ∼E7.5 Oct4 is required within a network that is altered relative to the pluripotency network. Oct4 is not required in extraembryonic tissue for these processes, but is required to maintain cell viability in the embryo and normal proliferation within the primitive streak. Impaired expansion of the primitive streak occurs coincident with Oct4 depletion ∼E7.5 and precedes deficient convergent extension which contributes to several aspects of the phenotype.


Vyšlo v časopise: Oct4 Is Required ∼E7.5 for Proliferation in the Primitive Streak. PLoS Genet 9(11): e32767. doi:10.1371/journal.pgen.1003957
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003957

Souhrn

Oct4 is a widely recognized pluripotency factor as it maintains Embryonic Stem (ES) cells in a pluripotent state, and, in vivo, prevents the inner cell mass (ICM) in murine embryos from differentiating into trophectoderm. However, its function in somatic tissue after this developmental stage is not well characterized. Using a tamoxifen-inducible Cre recombinase and floxed alleles of Oct4, we investigated the effect of depleting Oct4 in mouse embryos between the pre-streak and headfold stages, ∼E6.0–E8.0, when Oct4 is found in dynamic patterns throughout the embryonic compartment of the mouse egg cylinder. We found that depletion of Oct4 ∼E7.5 resulted in a severe phenotype, comprised of craniorachischisis, random heart tube orientation, failed turning, defective somitogenesis and posterior truncation. Unlike in ES cells, depletion of the pluripotency factors Sox2 and Oct4 after E7.0 does not phenocopy, suggesting that ∼E7.5 Oct4 is required within a network that is altered relative to the pluripotency network. Oct4 is not required in extraembryonic tissue for these processes, but is required to maintain cell viability in the embryo and normal proliferation within the primitive streak. Impaired expansion of the primitive streak occurs coincident with Oct4 depletion ∼E7.5 and precedes deficient convergent extension which contributes to several aspects of the phenotype.


Zdroje

1. NicholsJ, ZevnikB, AnastassiadisK, NiwaH, Klewe-NebeniusD, et al. (1998) Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 95: 379–391.

2. WangJ, RaoS, ChuJ, ShenX, LevasseurDN, et al. (2006) A protein interaction network for pluripotency of embryonic stem cells. Nature 444: 364–368.

3. LiangJ, WanM, ZhangY, GuP, XinH, et al. (2008) Nanog and Oct4 associate with unique transcriptional repression complexes in embryonic stem cells. Nat Cell Biol 10: 731–739.

4. PardoM, LangB, YuL, ProsserH, BradleyA, et al. (2010) An expanded Oct4 interaction network: implications for stem cell biology, development, and disease. Cell Stem Cell 6: 382–395.

5. ChenX, XuH, YuanP, FangF, HussM, et al. (2008) Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133: 1106–1117.

6. DownsKM (2008) Systematic localization of Oct-3/4 to the gastrulating mouse conceptus suggests manifold roles in mammalian development. Dev Dyn 237: 464–475.

7. ScholerHR, DresslerGR, BallingR, RohdewohldH, GrussP (1990) Oct-4: a germline-specific transcription factor mapping to the mouse t-complex. EMBO J 9: 2185–2195.

8. PesceM, WangX, WolgemuthDJ, ScholerH (1998) Differential expression of the Oct-4 transcription factor during mouse germ cell differentiation. Mech Dev 71: 89–98.

9. YoshimizuT, SugiyamaN, De FeliceM, YeomYI, OhboK, et al. (1999) Germline-specific expression of the Oct-4/green fluorescent protein (GFP) transgene in mice. Dev Growth Differ 41: 675–684.

10. BullejosM, KoopmanP (2004) Germ cells enter meiosis in a rostro-caudal wave during development of the mouse ovary. Mol Reprod Dev 68: 422–428.

11. BrennanJ, LuCC, NorrisDP, RodriguezTA, BeddingtonRS, et al. (2001) Nodal signalling in the epiblast patterns the early mouse embryo. Nature 411: 965–969.

12. WaldripWR, BikoffEK, HoodlessPA, WranaJL, RobertsonEJ (1998) Smad2 signaling in extraembryonic tissues determines anterior-posterior polarity of the early mouse embryo. Cell 92: 797–808.

13. FuhrmannG, ChungAC, JacksonKJ, HummelkeG, BaniahmadA, et al. (2001) Mouse germline restriction of Oct4 expression by germ cell nuclear factor. Dev Cell 1: 377–387.

14. TeoAK, ArnoldSJ, TrotterMW, BrownS, AngLT, et al. (2011) Pluripotency factors regulate definitive endoderm specification through eomesodermin. Genes Dev 25: 238–250.

15. NiwaH, SekitaY, Tsend-AyushE, GrutznerF (2008) Platypus Pou5f1 reveals the first steps in the evolution of trophectoderm differentiation and pluripotency in mammals. Evol Dev 10: 671–682.

16. FrankenbergS, PaskA, RenfreeMB (2010) The evolution of class V POU domain transcription factors in vertebrates and their characterisation in a marsupial. Dev Biol 337: 162–170.

17. BeddingtonRS (1983) Histogenetic and neoplastic potential of different regions of the mouse embryonic egg cylinder. J Embryol Exp Morphol 75: 189–204.

18. DamjanovI, SolterD, SkrebN (1971) Teratocarcinogenesis as related to the age of embryos grafted under the kidney capsule. Wilhelm Roux' Archiv für Entwicklungsmechanik der Organismen 167: 288–290.

19. OsornoR, TsakiridisA, WongF, CambrayN, EconomouC, et al. The developmental dismantling of pluripotency is reversed by ectopic Oct4 expression. Development 139: 2288–2298.

20. ThomsonM, LiuSJ, ZouLN, SmithZ, MeissnerA, et al. (2011) Pluripotency factors in embryonic stem cells regulate differentiation into germ layers. Cell 145: 875–889.

21. NiwaH, MiyazakiJ, SmithAG (2000) Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 24: 372–376.

22. ZeineddineD, PapadimouE, ChebliK, GinesteM, LiuJ, et al. (2006) Oct-3/4 dose dependently regulates specification of embryonic stem cells toward a cardiac lineage and early heart development. Dev Cell 11: 535–546.

23. ShimozakiK, NakashimaK, NiwaH, TagaT (2003) Involvement of Oct3/4 in the enhancement of neuronal differentiation of ES cells in neurogenesis-inducing cultures. Development 130: 2505–2512.

24. YuanH, CorbiN, BasilicoC, DaileyL (1995) Developmental-specific activity of the FGF-4 enhancer requires the synergistic action of Sox2 and Oct-3. Genes Dev 9: 2635–2645.

25. KehlerJ, TolkunovaE, KoschorzB, PesceM, GentileL, et al. (2004) Oct4 is required for primordial germ cell survival. EMBO Rep 5: 1078–1083.

26. HoL, RonanJL, WuJ, StaahlBT, ChenL, et al. (2009) An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency. Proc Natl Acad Sci U S A 106: 5181–5186.

27. van den BergDL, SnoekT, MullinNP, YatesA, BezstarostiK, et al. (2010) An Oct4-centered protein interaction network in embryonic stem cells. Cell Stem Cell 6: 369–381.

28. PesceM, ScholerHR (2001) Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells 19: 271–278.

29. SeiblerJ, ZevnikB, Kuter-LuksB, AndreasS, KernH, et al. (2003) Rapid generation of inducible mouse mutants. Nucleic Acids Res 31: e12.

30. CoppAJ, GreeneND, MurdochJN (2003) The genetic basis of mammalian neurulation. Nat Rev Genet 4: 784–793.

31. KalantryS, ManningS, HaubO, Tomihara-NewbergerC, LeeHG, et al. (2001) The amnionless gene, essential for mouse gastrulation, encodes a visceral-endoderm-specific protein with an extracellular cysteine-rich domain. Nat Genet 27: 412–416.

32. NagyA, GoczaE, DiazEM, PrideauxVR, IvanyiE, et al. (1990) Embryonic stem cells alone are able to support fetal development in the mouse. Development 110: 815–821.

33. McLarenA, MichieD (1956) Studies on the transfer of fertilized mouse eggs to the uterine foster-mothers I. Factors affecting the implantation and survival of native and transferred eggs. The Journal of Experimental Biology 33: 394–416.

34. TakashimaY, EraT, NakaoK, KondoS, KasugaM, et al. (2007) Neuroepithelial cells supply an initial transient wave of MSC differentiation. Cell 129: 1377–1388.

35. ParkEJ, SunX, NicholP, SaijohY, MartinJF, et al. (2008) System for tamoxifen-inducible expression of cre-recombinase from the Foxa2 locus in mice. Dev Dyn 237: 447–453.

36. FellerJ, SchneiderA, Schuster-GosslerK, GosslerA (2008) Noncyclic Notch activity in the presomitic mesoderm demonstrates uncoupling of somite compartmentalization and boundary formation. Genes Dev 22: 2166–2171.

37. NovakA, GuoC, YangW, NagyA, LobeCG (2000) Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genesis 28: 147–155.

38. HadjantonakisAK, PapaioannouVE (2004) Dynamic in vivo imaging and cell tracking using a histone fluorescent protein fusion in mice. BMC Biotechnol 4: 33.

39. TaranovaOV, MagnessST, FaganBM, WuY, SurzenkoN, et al. (2006) SOX2 is a dose-dependent regulator of retinal neural progenitor competence. Genes Dev 20: 1187–1202.

40. MasuiS, NakatakeY, ToyookaY, ShimosatoD, YagiR, et al. (2007) Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nat Cell Biol 9: 625–635.

41. AvilionAA, NicolisSK, PevnyLH, PerezL, VivianN, et al. (2003) Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev 17: 126–140.

42. KimJ, ChuJ, ShenX, WangJ, OrkinSH (2008) An extended transcriptional network for pluripotency of embryonic stem cells. Cell 132: 1049–1061.

43. MarsonA, LevineSS, ColeMF, FramptonGM, BrambrinkT, et al. (2008) Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134: 521–533.

44. KiermerV (2006) Embryos and biopsies on the ChIP-ing forecast. Nat Methods 3: 583.

45. NakatakeY, FukuiN, IwamatsuY, MasuiS, TakahashiK, et al. (2006) Klf4 cooperates with Oct3/4 and Sox2 to activate the Lefty1 core promoter in embryonic stem cells. Mol Cell Biol 26: 7772–7782.

46. HallJ, GuoG, WrayJ, EyresI, NicholsJ, et al. (2009) Oct4 and LIF/Stat3 additively induce Kruppel factors to sustain embryonic stem cell self-renewal. Cell Stem Cell 5: 597–609.

47. YiF, PereiraL, HoffmanJA, ShyBR, YuenCM, et al. (2011) Opposing effects of Tcf3 and Tcf1 control Wnt stimulation of embryonic stem cell self-renewal. Nat Cell Biol 13: 762–770.

48. SylvesterI, ScholerHR (1994) Regulation of the Oct-4 gene by nuclear receptors. Nucleic Acids Res 22: 901–911.

49. JungJS, JeeMK, ChoHT, ChoiJI, ImYB, et al. MBD6 is a direct target of Oct4 and controls the stemness and differentiation of adipose tissue-derived stem cells. Cell Mol Life Sci 70: 711–728.

50. MassagueJ, SeoaneJ, WottonD (2005) Smad transcription factors. Genes Dev 19: 2783–2810.

51. BeddingtonRS (1994) Induction of a second neural axis by the mouse node. Development 120: 613–620.

52. PrzemeckGK, HeinzmannU, BeckersJ, Hrabe de AngelisM (2003) Node and midline defects are associated with left-right development in Delta1 mutant embryos. Development 130: 3–13.

53. TamPP, BehringerRR (1997) Mouse gastrulation: the formation of a mammalian body plan. Mech Dev 68: 3–25.

54. ConlonFL, LyonsKM, TakaesuN, BarthKS, KispertA, et al. (1994) A primary requirement for nodal in the formation and maintenance of the primitive streak in the mouse. Development 120: 1919–1928.

55. ChuGC, DunnNR, AndersonDC, OxburghL, RobertsonEJ (2004) Differential requirements for Smad4 in TGFbeta-dependent patterning of the early mouse embryo. Development 131: 3501–3512.

56. TremblayKD, HoodlessPA, BikoffEK, RobertsonEJ (2000) Formation of the definitive endoderm in mouse is a Smad2-dependent process. Development 127: 3079–3090.

57. ZhouX, SasakiH, LoweL, HoganBL, KuehnMR (1993) Nodal is a novel TGF-beta-like gene expressed in the mouse node during gastrulation. Nature 361: 543–547.

58. FurtadoMB, SollowayMJ, JonesVJ, CostaMW, BibenC, et al. (2008) BMP/SMAD1 signaling sets a threshold for the left/right pathway in lateral plate mesoderm and limits availability of SMAD4. Genes Dev 22: 3037–3049.

59. CollignonJ, VarletI, RobertsonEJ (1996) Relationship between asymmetric nodal expression and the direction of embryonic turning. Nature 381: 155–158.

60. CandiaAF, WatabeT, HawleySH, OnichtchoukD, ZhangY, et al. (1997) Cellular interpretation of multiple TGF-beta signals: intracellular antagonism between activin/BVg1 and BMP-2/4 signaling mediated by Smads. Development 124: 4467–4480.

61. TzouanacouE, WegenerA, WymeerschFJ, WilsonV, NicolasJF (2009) Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis. Dev Cell 17: 365–376.

62. MiyagiS, MasuiS, NiwaH, SaitoT, ShimazakiT, et al. (2008) Consequence of the loss of Sox2 in the developing brain of the mouse. FEBS Lett 582: 2811–2815.

63. SchoenwolfGC, DesmondME (1984) Neural tube occlusion precedes rapid brain enlargement. J Exp Zool 230: 405–407.

64. MorrisonGM, BrickmanJM (2006) Conserved roles for Oct4 homologues in maintaining multipotency during early vertebrate development. Development 133: 2011–2022.

65. SnirM, OfirR, EliasS, FrankD (2006) Xenopus laevis POU91 protein, an Oct3/4 homologue, regulates competence transitions from mesoderm to neural cell fates. EMBO J 25: 3664–3674.

66. ChapmanDL, PapaioannouVE (1998) Three neural tubes in mouse embryos with mutations in the T-box gene Tbx6. Nature 391: 695–697.

67. YoshikawaY, FujimoriT, McMahonAP, TakadaS (1997) Evidence that absence of Wnt-3a signaling promotes neuralization instead of paraxial mesoderm development in the mouse. Dev Biol 183: 234–242.

68. WangY, BlellochR (2009) Cell cycle regulation by MicroRNAs in embryonic stem cells. Cancer Res 69: 4093–4096.

69. KimJ, WooAJ, ChuJ, SnowJW, FujiwaraY, et al. (2010) A Myc network accounts for similarities between embryonic stem and cancer cell transcription programs. Cell 143: 313–324.

70. MateyakMK, ObayaAJ, SedivyJM (1999) c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points. Mol Cell Biol 19: 4672–4683.

71. ReimG, MizoguchiT, StainierDY, KikuchiY, BrandM (2004) The POU domain protein spg (pou2/Oct4) is essential for endoderm formation in cooperation with the HMG domain protein casanova. Dev Cell 6: 91–101.

72. WoodSA, AllenND, RossantJ, AuerbachA, NagyA (1993) Non-injection methods for the production of embryonic stem cell-embryo chimaeras. Nature 365: 87–89.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 11
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#