#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

SOX2 Co-Occupies Distal Enhancer Elements with Distinct POU Factors in ESCs and NPCs to Specify Cell State


SOX2 is a master regulator of both pluripotent embryonic stem cells (ESCs) and multipotent neural progenitor cells (NPCs); however, we currently lack a detailed understanding of how SOX2 controls these distinct stem cell populations. Here we show by genome-wide analysis that, while SOX2 bound to a distinct set of gene promoters in ESCs and NPCs, the majority of regions coincided with unique distal enhancer elements, important cis-acting regulators of tissue-specific gene expression programs. Notably, SOX2 bound the same consensus DNA motif in both cell types, suggesting that additional factors contribute to target specificity. We found that, similar to its association with OCT4 (Pou5f1) in ESCs, the related POU family member BRN2 (Pou3f2) co-occupied a large set of putative distal enhancers with SOX2 in NPCs. Forced expression of BRN2 in ESCs led to functional recruitment of SOX2 to a subset of NPC-specific targets and to precocious differentiation toward a neural-like state. Further analysis of the bound sequences revealed differences in the distances of SOX and POU peaks in the two cell types and identified motifs for additional transcription factors. Together, these data suggest that SOX2 controls a larger network of genes than previously anticipated through binding of distal enhancers and that transitions in POU partner factors may control tissue-specific transcriptional programs. Our findings have important implications for understanding lineage specification and somatic cell reprogramming, where SOX2, OCT4, and BRN2 have been shown to be key factors.


Vyšlo v časopise: SOX2 Co-Occupies Distal Enhancer Elements with Distinct POU Factors in ESCs and NPCs to Specify Cell State. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003288
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003288

Souhrn

SOX2 is a master regulator of both pluripotent embryonic stem cells (ESCs) and multipotent neural progenitor cells (NPCs); however, we currently lack a detailed understanding of how SOX2 controls these distinct stem cell populations. Here we show by genome-wide analysis that, while SOX2 bound to a distinct set of gene promoters in ESCs and NPCs, the majority of regions coincided with unique distal enhancer elements, important cis-acting regulators of tissue-specific gene expression programs. Notably, SOX2 bound the same consensus DNA motif in both cell types, suggesting that additional factors contribute to target specificity. We found that, similar to its association with OCT4 (Pou5f1) in ESCs, the related POU family member BRN2 (Pou3f2) co-occupied a large set of putative distal enhancers with SOX2 in NPCs. Forced expression of BRN2 in ESCs led to functional recruitment of SOX2 to a subset of NPC-specific targets and to precocious differentiation toward a neural-like state. Further analysis of the bound sequences revealed differences in the distances of SOX and POU peaks in the two cell types and identified motifs for additional transcription factors. Together, these data suggest that SOX2 controls a larger network of genes than previously anticipated through binding of distal enhancers and that transitions in POU partner factors may control tissue-specific transcriptional programs. Our findings have important implications for understanding lineage specification and somatic cell reprogramming, where SOX2, OCT4, and BRN2 have been shown to be key factors.


Zdroje

1. YoungRA (2011) Control of the Embryonic Stem Cell State. Cell 144: 940–954.

2. HolmbergJ, PerlmannT (2012) Maintaining Differentiated Cellular Identity. Nature Reviews Genetics 13: 429–439.

3. AvilionAA, NicolisSK, PevnyLH, PerezL, VivianN, et al. (2003) Multipotent Cell Lineages in Early Mouse Development Depend on Sox2 Function. Genes & Development 17: 126.

4. MasuiS, NakatakeY, ToyookaY, ShimosatoD, YagiR, et al. (2007) Pluripotency Governed by Sox2 Via Regulation of Oct3/4 Expression in Mouse Embryonic Stem Cells. Nature Cell Biology 9: 625–635.

5. MiyagiS, MasuiS, NiwaH, SaitoT, ShimazakiT, et al. (2008) Consequence of the Loss of Sox2 in the Developing Brain of the Mouse. FEBS Letters 582: 2811–2815.

6. FavaroR, ValottaM, FerriALM, LatorreE, MarianiJ, et al. (2009) Hippocampal Development and Neural Stem Cell Maintenance Require Sox2-Dependent Regulation of Shh. Nature Neuroscience 12: 1248–1256.

7. KamachiY, SockanathanS, LiuQ, BreitmanM, Lovell-BadgeR, et al. (1995) Involvement of Sox Proteins in Lens-Specific Activation of Crystallin Genes. The EMBO Journal 14: 3510.

8. KiernanAE, PellingAL, LeungKKH, TangASP, BellDM, et al. (2005) Sox2 Is Required for Sensory Organ Development in the Mammalian Inner Ear. Nature 434: 1031–1035.

9. OkuboT, PevnyLH, HoganBLM (2006) Sox2 Is Required for Development of Taste Bud Sensory Cells. Genes & Development 20: 2654.

10. GontanC, de MunckA, VermeijM, GrosveldF, TibboelD, et al. (2008) Sox2 Is Important for Two Crucial Processes in Lung Development: Branching Morphogenesis and Epithelial Cell Differentiation. Developmental Biology 317: 296–309.

11. DomyanET, FerrettiE, ThrockmortonK, MishinaY, NicolisSK, et al. (2011) Signaling through Bmp Receptors Promotes Respiratory Identity in the Foregut Via Repression of Sox2. Development 138: 971–981.

12. ArnoldK, SarkarA, YramMA, PoloJM, BronsonR, et al. (2011) Sox2+ Adult Stem and Progenitor Cells Are Important for Tissue Regeneration and Survival of Mice. Cell Stem Cell 9: 317–329.

13. BoyerLA, LeeTI, ColeMF, JohnstoneSE, LevineSS, et al. (2005) Core Transcriptional Regulatory Circuitry in Human Embryonic Stem Cells. Cell 122: 947–956.

14. 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.

15. LohYH, WuQ, ChewJL, VegaVB, ZhangW, et al. (2006) The Oct4 and Nanog Transcription Network Regulates Pluripotency in Mouse Embryonic Stem Cells. Nature Genetics 38: 431–440.

16. 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.

17. BergslandM, RamsköldD, ZaouterC, KlumS, SandbergR, et al. (2011) Sequentially Acting Sox Transcription Factors in Neural Lineage Development. Genes & Development 25: 2453–2464.

18. LiberD, DomaschenzR, HolmqvistPH, MazzarellaL, GeorgiouA, et al. (2010) Epigenetic Priming of a Pre-B Cell-Specific Enhancer through Binding of Sox2 and Foxd3 at the ESC Stage. Cell Stem Cell 7: 114–126.

19. TakahashiK, TanabeK, OhnukiM, NaritaM, IchisakaT, et al. (2007) Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 131: 861–872.

20. TakahashiK, YamanakaS (2006) Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 126: 663–676.

21. WernigM, MeissnerA, ForemanR, BrambrinkT, KuM, et al. (2007) In Vitro Reprogramming of Fibroblasts into a Pluripotent Es-Cell-Like State. Nature 448: 318–324.

22. BuganimY, FaddahDA, ChengAW, ItskovichE, MarkoulakiS, et al. (2012) Single-Cell Expression Analyses During Cellular Reprogramming Reveal an Early Stochastic and a Late Hierarchic Phase. Cell 150: 1209–1222.

23. TempleS, Alvarez-BuyllaA (1999) Stem Cells in the Adult Mammalian Central Nervous System. Current Opinion in Neurobiology 9: 135–141.

24. TempleS (2001) The Development of Neural Stem Cells. Nature 414: 112–117.

25. SuhH, ConsiglioA, RayJ, SawaiT, D'AmourKA, et al. (2007) In Vivo Fate Analysis Reveals the Multipotent and Self-Renewal Capacities of Sox2+ Neural Stem Cells in the Adult Hippocampus. Cell Stem Cell 1: 515–528.

26. BylundM, AnderssonE, NovitchBG, MuhrJ (2003) Vertebrate Neurogenesis Is Counteracted by Sox1–3 Activity. Nature Neuroscience 6: 1162–1168.

27. GrahamV, KhudyakovJ, EllisP, PevnyL (2003) Sox2 Functions to Maintain Neural Progenitor Identity. Neuron 39: 749–765.

28. Bani-YaghoubM, TremblayRG, LeiJX, ZhangD, ZurakowskiB, et al. (2006) Role of Sox2 in the Development of the Mouse Neocortex. Developmental Biology 295: 52–66.

29. LeN, NagarajanR, WangJYT, ArakiT, SchmidtRE, et al. (2005) Analysis of Congenital Hypomyelinating Egr2lo/Lo Nerves Identifies Sox2 as an Inhibitor of Schwann Cell Differentiation and Myelination. Proc Natl Acad Sci USA 102: 2596.

30. EngelenE, AkinciU, BryneJC, HouJ, GontanC, et al. (2011) Sox2 Cooperates with Chd7 to Regulate Genes That Are Mutated in Human Syndromes. Nature Genetics 43: 607–611.

31. BueckerC, WysockaJ (2012) Enhancers as Information Integration Hubs in Development: Lessons from Genomics. Trends in Genetics 6: 276–284.

32. BulgerM, GroudineM (2011) Functional and Mechanistic Diversity of Distal Transcription Enhancers. Cell 144: 327–339.

33. OngCT, CorcesVG (2011) Enhancer Function: New Insights into the Regulation of Tissue-Specific Gene Expression. Nature Reviews Genetics 12: 283–293.

34. CreyghtonMP, ChengAW, WelsteadGG, KooistraT, CareyBW, et al. (2010) Histone H3k27ac Separates Active from Poised Enhancers and Predicts Developmental State. Proc Natl Acad Sci USA 107: 21931.

35. HeintzmanND, HonGC, HawkinsRD, KheradpourP, StarkA, et al. (2009) Histone Modifications at Human Enhancers Reflect Global Cell-Type-Specific Gene Expression. Nature 459: 108–112.

36. HeintzmanND, StuartRK, HonG, FuY, ChingCW, et al. (2007) Distinct and Predictive Chromatin Signatures of Transcriptional Promoters and Enhancers in the Human Genome. Nature Genetics 39: 311–318.

37. Rada-IglesiasA, BajpaiR, SwigutT, BrugmannSA, FlynnRA, et al. (2010) A Unique Chromatin Signature Uncovers Early Developmental Enhancers in Humans. Nature 470: 279–283.

38. TanakaS, KamachiY, TanouchiA, HamadaH, JingN, et al. (2004) Interplay of Sox and Pou Factors in Regulation of the Nestin Gene in Neural Primordial Cells. Molecular and Cellular Biology 24: 8834.

39. ChewJL, LohYH, ZhangW, ChenX, TamWL, et al. (2005) Reciprocal Transcriptional Regulation of Pou5f1 and Sox2 Via the Oct4/Sox2 Complex in Embryonic Stem Cells. Molecular and Cellular Biology 25: 6031–6046.

40. JinZ, LiuL, BianW, ChenY, XuG, et al. (2009) Different Transcription Factors Regulate Nestin Gene Expression During P19 Cell Neural Differentiation and Central Nervous System Development. Journal of Biological Chemistry 284: 8160–8173.

41. KondohH, KamachiY (2010) Sox-Partner Code for Cell Specification: Regulatory Target Selection and Underlying Molecular Mechanisms. The International Journal of Biochemistry and Cell Biology 42: 391–399.

42. DaileyL, BasilicoC (2001) Coevolution of Hmg Domains and Homeodomains and the Generation of Transcriptional Regulation by Sox/Pou Complexes. Journal of Cellular Physiology 186: 315–328.

43. OkabeS, Forsberg-NilssonK, SpiroAC, SegalM, McKayRDG (1996) Development of Neuronal Precursor Cells and Functional Postmitotic Neurons from Embryonic Stem Cells in Vitro. Mechanisms of Development 59: 89–102.

44. MitsuiK, TokuzawaY, ItohH, SegawaK, MurakamiM, et al. (2003) The Homeoprotein Nanog Is Required for Maintenance of Pluripotency in Mouse Epiblast and Es Cells. Cell 113: 631–642.

45. ChambersI, ColbyD, RobertsonM, NicholsJ, LeeS, et al. (2003) Functional Expression Cloning of Nanog, a Pluripotency Sustaining Factor in Embryonic Stem Cells. Cell 113: 643–655.

46. Schneider-MaunouryS, TopilkoP, SeitandouT, LeviG, Cohen-TannoudjiM, et al. (1993) Disruption of Krox-20 Results in Alteration of Rhombomeres 3 and 5 in the Developing Hindbrain. Cell 75: 1199–1214.

47. ShamMH, VesqueC, NonchevS, MarshallH, FrainM, et al. (1993) The Zinc Finger Gene Krox20 Regulates Hoxb2 (Hox2.8) During Hindbrain Segmentation. Cell 72: 183–196.

48. SugoN, OshiroH, TakemuraM, KobayashiT, KohnoY, et al. (2010) Nucleocytoplasmic Translocation of Hdac9 Regulates Gene Expression and Dendritic Growth in Developing Cortical Neurons. European Journal of Neuroscience 31: 1521–1532.

49. ShinJ, BossenzM, ChungY, MaH, ByronM, et al. (2010) Maternal Rnf12/Rlim Is Required for Imprinted X-Chromosome Inactivation in Mice. Nature 467: 977–981.

50. OstendorffHP, TursunB, CornilsK, SchluterA, DrungA, et al. (2006) Dynamic Expression of Lim Cofactors in the Developing Mouse Neural Tube. Developmental Dynamics 235: 786–791.

51. GrabowskiP (2011) Alternative Splicing Takes Shape During Neuronal Development. Current Opinion in Genetics & Development 4: 388–94.

52. BoutzPL, StoilovP, LiQ, LinCH, ChawlaG, et al. (2007) A Post-Transcriptional Regulatory Switch in Polypyrimidine Tract-Binding Proteins Reprograms Alternative Splicing in Developing Neurons. Genes & Development 21: 1636.

53. WamstadJA, AlexanderJM, TrutyRM, ShrikumarA, LiF, et al. (2012) Dynamic and Coordinated Epigenetic Regulation of Developmental Transitions in the Cardiac Lineage. Cell 151: 206–220.

54. ErnstJ, KheradpourP, MikkelsenTS, ShoreshN, WardLD, et al. (2011) Mapping and Analysis of Chromatin State Dynamics in Nine Human Cell Types. Nature 473: 43–49.

55. ZentnerGE, TesarPJ, ScacheriPC (2011) Epigenetic Signatures Distinguish Multiple Classes of Enhancers with Distinct Cellular Functions. Genome Research 21: 1273–1283.

56. JiangJ, ChanYS, LohYH, CaiJ, TongGQ, et al. (2008) A Core Klf Circuitry Regulates Self-Renewal of Embryonic Stem Cells. Nature Cell Biology 10: 353–360.

57. TzatzalosE, SmithSM, DohST, HaoH, LiY, et al. (2012) A Cis-Element in the Notch1 Locus Is Involved in the Regulation of Gene Expression in Interneuron Progenitors. Developmental Biology 372 (2)

217–28.

58. TaranovaOV, MagnessST, FaganBM, WuY, SurzenkoN, et al. (2006) Sox2 Is a Dose-Dependent Regulator of Retinal Neural Progenitor Competence. Genes & Development 20: 1187–1202.

59. KondohH, KamachiY (2010) Sox-Partner Code for Cell Specification: Regulatory Target Selection and Underlying Molecular Mechanisms. The International Journal of Biochemistry & Cell Biology 42: 391–399.

60. YuanH, CorbiN, BasilicoC, DaileyL (1995) Developmental-Specific Activity of the Fgf-4 Enhancer Requires the Synergistic Action of Sox2 and Oct-3. Genes & Development 9: 2635.

61. CollignonJ, SockanathanS, HackerA, Cohen-TannoudjiM, NorrisD, et al. (1996) A Comparison of the Properties of Sox-3 with Sry and Two Related Genes, Sox-1 and Sox-2. Development 122: 509.

62. KamachiY, CheahKSE, KondohH (1999) Mechanism of Regulatory Target Selection by the Sox High-Mobility-Group Domain Proteins as Revealed by Comparison of Sox1/2/3 and Sox9. Molecular and Cellular Biology 19: 107.

63. Salmon-DivonM, DvingeH, TammojaK, BertoneP (2010) Peakanalyzer: Genome-Wide Annotation of Chromatin Binding and Modification Loci. BMC Bioinformatics 11: 415.

64. CatenaR, TiveronC, RonchiA, PortaS, FerriA, et al. (2004) Conserved Pou Binding DNA Sites in the Sox2 Upstream Enhancer Regulate Gene Expression in Embryonic and Neural Stem Cells. Journal of Biological Chemistry 279: 41846–41857.

65. MiyagiS, NishimotoM, SaitoT, NinomiyaM, SawamotoK, et al. (2006) The Sox2 Regulatory Region 2 Functions as a Neural Stem Cell-Specific Enhancer in the Telencephalon. Journal of Biological Chemistry 281: 13374–13381.

66. HeX, TreacyMN, SimmonsDM, IngrahamHA, SwansonLW, et al. (1989) Expression of a Large Family of Pou-Domain Regulatory Genes in Mammalian Brain Development. Nature 340: 35–41.

67. HaraY, RovescalliAC, KimY, NirenbergM (1992) Structure and Evolution of Four Pou Domain Genes Expressed in Mouse Brain. Proc Natl Acad Sci USA 89: 3280.

68. McEvillyRJ, de DiazMO, SchonemannMD, HooshmandF, RosenfeldMG (2002) Transcriptional Regulation of Cortical Neuron Migration by Pou Domain Factors. Science 295: 1528.

69. SugitaniY, NakaiS, MinowaO, NishiM, JishageK, et al. (2002) Brn-1 and Brn-2 Share Crucial Roles in the Production and Positioning of Mouse Neocortical Neurons. Genes & Development 16: 1760.

70. LujanE, ChandaS, AhleniusH, SüdhofTC, WernigM (2012) Direct Conversion of Mouse Fibroblasts to Self-Renewing, Tripotent Neural Precursor Cells. Proc Natl Acad Sci USA 109: 2527–2532.

71. StaudtLM, SinghH, SenR, WirthT, SharpPA, et al. (1986) A Lymphoid-Specific Protein Binding to the Octamer Motif of Immunoglobulin Genes. Nature 323: 640–643.

72. PhillipsK, LuisiB (2000) The Virtuoso of Versatility: Pou Proteins That Flex to Fit1. Journal of Molecular Biology 302: 1023–1039.

73. KleinjanDA, SeawrightA, ChildsAJ, van HeyningenV (2004) Conserved Elements in Pax6 Intron 7 Involved in (Auto)Regulation and Alternative Transcription. Developmental Biology 265: 462–477.

74. FriedliM, BardeI, ArcangeliM, VerpS, QuazzolaA, et al. (2010) A Systematic Enhancer Screen Using Lentivector Transgenesis Identifies Conserved and Non-Conserved Functional Elements at the Olig1 and Olig2 Locus. PLoS ONE 5: e15741 doi:10.1371/journal.pone.0015741.

75. Verma-KurvariS, SavageT, SmithD, JohnsonJE (1998) Multiple Elements Regulate Mash1 Expression in the Developing Cns. Developmental Biology 197: 106–116.

76. RoseMF, AhmadKA, ThallerC, ZoghbiHY (2009) Excitatory Neurons of the Proprioceptive, Interoceptive, and Arousal Hindbrain Networks Share a Developmental Requirement for Math1. Proc Natl Acad Sci USA 106: 22462–22467.

77. LaiHC, KlischTJ, RobertsR, ZoghbiHY, JohnsonJE (2011) In Vivo Neuronal Subtype-Specific Targets of Atoh1 (Math1) in Dorsal Spinal Cord. The Journal of Neuroscience 31: 10859–10871.

78. HondaT, KobayashiK, MikoshibaK, NakajimaK (2011) Regulation of Cortical Neuron Migration by the Reelin Signaling Pathway. Neurochemical Research 36: 1270–1279.

79. AndreaeLC, PeukertD, LumsdenA, GilthorpeJD (2007) Analysis of Lrrn1 Expression and Its Relationship to Neuromeric Boundaries During Chick Neural Development. Neural Development 2: 22.

80. TaguchiA, WanakaA, MoriT, MatsumotoK, ImaiY, et al. (1996) Molecular Cloning of Novel Leucine-Rich Repeat Proteins and Their Expression in the Developing Mouse Nervous System. Brain Res Mol Brain Res 35: 31–40.

81. SanoY, Syuzo-TakabatakeA, NakayaT, SaitoY, TomitaS, et al. (2006) Enhanced Amyloidogenic Metabolism of the Amyloid Beta-Protein Precursor in the X11l-Deficient Mouse Brain. Journal of Biological Chemistry 281: 37853–37860.

82. BotquinV, HessH, FuhrmannG, AnastassiadisC, GrossMK, et al. (1998) New Pou Dimer Configuration Mediates Antagonistic Control of an Osteopontin Preimplantation Enhancer by Oct-4 and Sox-2. Genes & Development 12: 2073–2090.

83. NishimotoM, FukushimaA, OkudaA, MuramatsuM (1999) The Gene for the Embryonic Stem Cell Coactivator Utf1 Carries a Regulatory Element Which Selectively Interacts with a Complex Composed of Oct-3/4 and Sox-2. Molecular and Cellular Biology 19: 5453–5465.

84. AmbrosettiDC, SchölerHR, DaileyL, BasilicoC (2000) Modulation of the Activity of Multiple Transcriptional Activation Domains by the DNA Binding Domains Mediates the Synergistic Action of Sox2 and Oct-3 on the Fibroblast Growth Factor-4 enhancer. Journal of Biological Chemistry 275: 23387.

85. ReményiA, LinsK, NissenLJ, ReinboldR, SchölerHR, et al. (2003) Crystal Structure of a Pou/Hmg/DNA Ternary Complex Suggests Differential Assembly of Oct4 and Sox2 on Two Enhancers. Genes & Development 17: 2048–2059.

86. JauchR, AksoyI, HutchinsAP, NgCKL, TianXF, et al. (2011) Conversion of Sox17 into a Pluripotency Reprogramming Factor by Reengineering Its Association with Oct4 on DNA. Stem Cells 29: 940–951.

87. AmbrosettiDC, BasilicoC, DaileyL (1997) Synergistic Activation of the Fibroblast Growth Factor 4 Enhancer by Sox2 and Oct-3 Depends on Protein-Protein Interactions Facilitated by a Specific Spatial Arrangement of Factor Binding Sites. Molecular and Cellular Biology 17: 6321.

88. FerrarisL, StewartAP, KangJ, DeSimoneAM, GemberlingM, et al. (2011) Combinatorial Binding of Transcription Factors in the Pluripotency Control Regions of the Genome. Genome Research 21: 1055–1064.

89. NgCKL, LiNX, CheeS, PrabhakarS, KolatkarPR, et al. (2012) Deciphering the Sox-Oct Partner Code by Quantitative Cooperativity Measurements. Nucleic Acids Research

90. RemenyiA, ScholerHR, WilmannsM (2004) Combinatorial Control of Gene Expression. Nature Structural and Molecular Biology 11: 812–815.

91. Rochette-EglyC, GermainP (2009) Dynamic and Combinatorial Control of Gene Expression by Nuclear Retinoic Acid Receptors (RARs). Nuclear Receptor Signalling 7: e005.

92. SundrudMS, NolanMA (2010) Synergistic and Combinatorial Control of T Cell Activation and Differentiation by Transcription Factors. Curr Opin Immunol 22: 286–292.

93. MichelD (2010) How Transcription Factors Can Adjust the Gene Expression Floodgates. Progress in Biophysics and Molecular Biology 102: 16–37.

94. KimJ, ChuJ, ShenX, WangJ, OrkinSH (2008) An Extended Transcriptional Network for Pluripotency of Embryonic Stem Cells. Cell 132: 1049–1061.

95. YangJ, ChaiL, FowlesTC, AlipioZ, XuD, et al. (2008) Genome-Wide Analysis Reveals Sall4 to Be a Major Regulator of Pluripotency in Murine-Embryonic Stem Cells. Proc Natl Acad Sci USA 105: 19756–19761.

96. ColeMF, JohnstoneSE, NewmanJJ, KageyMH, YoungRA (2008) Tcf3 Is an Integral Component of the Core Regulatory Circuitry of Embryonic Stem Cells. Genes & Development 22: 746.

97. das NevesL, DuchalaCS, Tolentino-SilvaF, HaxhiuMA, ColmenaresC, et al. (1999) Disruption of the Murine Nuclear Factor I-a Gene (Nfia) Results in Perinatal Lethality, Hydrocephalus, and Agenesis of the Corpus Callosum. Proc Natl Acad Sci USA 96: 11946–11951.

98. BaasD, MeinielA, BenadibaC, BonnafeE, MeinielO, et al. (2006) A Deficiency in Rfx3 Causes Hydrocephalus Associated with Abnormal Differentiation of Ependymal Cells. Europena Journal Neuroscience 24: 1020–1030.

99. AshiqueAM, ChoeY, KarlenM, MaySR, PhamluongK, et al. (2009) The Rfx4 Transcription Factor Modulates Shh Signaling by Regional Control of Ciliogenesis. Science Signalling 2: ra70.

100. AlcantaraS, FrisenJ, del RioJA, SorianoE, BarbacidM, et al. (1997) Trkb Signaling Is Required for Postnatal Survival of Cns Neurons and Protects Hippocampal and Motor Neurons from Axotomy-Induced Cell Death. The Journal of Neuroscience 17: 3623–3633.

101. PostigoA, CalellaAM, FritzschB, KnipperM, KatzD, et al. (2002) Distinct Requirements for Trkb and Trkc Signaling in Target Innervation by Sensory Neurons. Genes & Development 16: 633–645.

102. O'HareMJ, KushwahaN, ZhangY, AleyasinH, CallaghanSM, et al. (2005) Differential Roles of Nuclear and Cytoplasmic Cyclin-Dependent Kinase 5 in Apoptotic and Excitotoxic Neuronal Death. The Journal of Neuroscience 25: 8954–8966.

103. RochetJC (2007) Novel Therapeutic Strategies for the Treatment of Protein-Misfolding Diseases. Expert Rev Mol Med 9: 1–34.

104. GrittiA, ParatiE, CovaL, FrolichsthalP, GalliR, et al. (1996) Multipotential Stem Cells from the Adult Mouse Brain Proliferate and Self-Renew in Response to Basic Fibroblast Growth Factor. The Journal of Neuroscience 16: 1091–1100.

105. TropepeV, SibiliaM, CirunaBG, RossantJ, WagnerEF, et al. (1999) Distinct Neural Stem Cells Proliferate in Response to Egf and Fgf in the Developing Mouse Telencephalon. Developmental Biology 208: 166–188.

106. QianX, ShenQ, GoderieSK, HeW, CapelaA, et al. (2000) A Programmed Sequence of Neuron and Glial Cell Production from Isolated Murine Cortical Stem Cells. Neuron 28: 69–80.

107. YasuharaN, ShibazakiN, TanakaS, NagaiM, KamikawaY, et al. (2006) Triggering Neural Differentiation of ES Cells by Subtype Switching of Importin-A. Nature Cell Biology 9: 72–79.

108. Iwafuchi-DoiM, YoshidaY, OnichtchoukD, LeichsenringM, DrieverW, et al. (2010) The Pou5f1/Pou3f-Dependent but SoxB-Independent Regulation of Conserved Enhancer N2 Initiates Sox2 Expression During Epiblast to Neural Plate Stages in Vertebrates. Developmental Biology 352 (2)

354–66.

109. KimJB, SebastianoV, WuG, Araúzo-BravoMJ, SasseP, et al. (2009) Oct4-Induced Pluripotency in Adult Neural Stem Cells. Cell 136: 411–419.

110. KuhlbrodtK, HerbarthB, SockE, EnderichJ, Hermans-BorgmeyerI, et al. (1998) Cooperative Function of Pou Proteins and Sox Proteins in Glial Cells. Journal of Biological Chemistry 273: 16050.

111. GhislainJ, CharnayP (2006) Control of Myelination in Schwann Cells: A Krox20 Cis-Regulatory Element Integrates Oct6, Brn2 and Sox10 Activities. EMBO Rep 7: 52–58.

112. 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. Molecular and Cellular Biology 26: 7772–7782.

113. ReiprichS, KrieschJ, SchreinerS, WegnerM (2010) Activation of Krox20 Gene Expression by Sox10 in Myelinating Schwann Cells. Journal of Neurochemistry 112: 744–754.

114. NowlingTK, JohnsonLR, WiebeMS, RizzinoA (2000) Identification of the Transactivation Domain of the Transcription Factor Sox-2 and an Associated Co-Activator. Journal of Biological Chemistry 275: 3810–3818.

115. WilliamsDCJr, CaiM, CloreGM (2004) Molecular Basis for Synergistic Transcriptional Activation by Oct1 and Sox2 Revealed from the Solution Structure of the 42-Kda Oct1· Sox2· Hoxb1-DNA Ternary Transcription Factor Complex. Journal of Biological Chemistry 279: 1449–1457.

116. Maniatis T, Falvo J, Kim T, Kim T, Lin C, et al.. Structure and Function of the Interferon-B Enhanceosome; 1998. Cold Spring Harbor Laboratory Press. pp. 609–620.

117. WangJ, RaoS, ChuJ, ShenX, LevasseurDN, et al. (2006) A Protein Interaction Network for Pluripotency of Embryonic Stem Cells. Nature 444: 364–368.

118. LiangJ, WanM, ZhangY, GuP, XinH, et al. (2008) Nanog and Oct4 Associate with Unique Transcriptional Repression Complexes in Embryonic Stem Cells. Nature Cell Biology 10: 731–739.

119. Van Den BergDLC, SnoekT, MullinNP, YatesA, BezstarostiK, et al. (2010) An Oct4-Centered Protein Interaction Network in Embryonic Stem Cells. Cell Stem Cell 6: 369–381.

120. ChambersI, TomlinsonSR (2009) The Transcriptional Foundation of Pluripotency. Development 136: 2311–2322.

121. Steele-PerkinsG, PlachezC, ButzKG, YangG, BachurskiCJ, et al. (2005) The Transcription Factor Gene Nfib Is Essential for Both Lung Maturation and Brain Development. Molecular and Cellular Biology 25: 685–698.

122. PiperM, BarryG, HawkinsJ, MasonS, LindwallC, et al. (2010) NfIA Controls Telencephalic Progenitor Cell Differentiation through Repression of the Notch Effector Hes1. The Journal of Neuroscience 30: 9127–9139.

123. CampbellCE, PiperM, PlachezC, YehYT, BaizerJS, et al. (2008) The Transcription Factor NfIX Is Essential for Normal Brain Development. BMC Developmental Biology 8: 52.

124. PlachezC, LindwallC, SunnN, PiperM, MoldrichRX, et al. (2008) Nuclear Factor I Gene Expression in the Developing Forebrain. Journal of Computational Neurology 508: 385–401.

125. BlackshearPJ, GravesJP, StumpoDJ, CobosI, RubensteinJLR, 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.

126. OkamotoK, WakamiyaM, NojiS, KoyamaE, TaniguchiS, et al. (1993) A Novel Class of Murine Pou Gene Predominantly Expressed in Central Nervous System. Journal of Biological Chemistry 268: 7449–7457.

127. AndersenB, SchonemannM, PearseRV, JenneK, SugarmanJ, et al. (1993) Brn-5 Is a Divergent Pou Domain Factor Highly Expressed in Layer Iv of the Neocortex. Journal of Biological Chemistry 268: 23390.

128. CuiH, BulleitRF (1997) Expression of the Pou Transcription Factor Brn-5 Inhibits Proliferation of Ng108-15 Cells. Biochemical and Biophysical Research Communications 236: 693–696.

129. CuiH, BulleitRF (1998) Expression of the POU Transcription Factor Brn‐5 Is an Early Event in the Terminal Differentiation of CNS Neurons. Journal of Neuroscience Research 52: 625–632.

130. WuR, JurekM, SundarababuS, WeinsteinDE (2001) The Pou Gene Brn-5 Is Induced by Neuregulin and Is Restricted to Myelinating Schwann Cells. Molecular and Cellular Neuroscience 17: 683–695.

131. LeeTI, JohnstoneSE, YoungRA (2006) Chromatin Immunoprecipitation and Microarray-Based Analysis of Protein Location. Nature Protocols 1: 729–748.

132. MikkelsenTS, KuM, JaffeDB, IssacB, LiebermanE, et al. (2007) Genome-Wide Maps of Chromatin State in Pluripotent and Lineage-Committed Cells. Nature 448: 553–560.

133. BilodeauS, KageyMH, FramptonGM, RahlPB, YoungRA (2009) Setdb1 Contributes to Repression of Genes Encoding Developmental Regulators and Maintenance of ES Cell State. Genes & Development 23: 2484.

134. McLeanCY, BristorD, HillerM, ClarkeSL, SchaarBT, et al. (2010) Great Improves Functional Interpretation of Cis-Regulatory Regions. Nature Biotechnology 28: 495–501.

135. BaileyTL, ElkanC (1994) Fitting a Mixture Model by Expectation Maximization to Discover Motifs in Biopolymers. Proc Int Conf Intell Syst Mol Biol 2: 28–36.

136. BeardC, HochedlingerK, PlathK, WutzA, JaenischR (2006) Efficient Method to Generate Single-Copy Transgenic Mice by Site-Specific Integration in Embryonic Stem Cells. Genesis 44: 23–28.

137. YingQL, StavridisM, GriffithsD, LiM, SmithA (2003) Conversion of Embryonic Stem Cells into Neuroectodermal Precursors in Adherent Monoculture. Nature Biotechnology 21: 183–186.

138. WingenderE, DietzeP, KarasH, KnuppelR (1996) Transfac: A Database on Transcription Factors and Their DNA Binding Sites. Nucleic Acids Research 24: 238–241.

139. GordonDB, NekludovaL, McCallumS, FraenkelE (2005) Tamo: A Flexible, Object-Oriented Framework for Analyzing Transcriptional Regulation Using DNA-Sequence Motifs. Bioinformatics 21: 3164–3165.

140. MacisaacKD, GordonDB, NekludovaL, OdomDT, SchreiberJ, et al. (2006) A Hypothesis-Based Approach for Identifying the Binding Specificity of Regulatory Proteins from Chromatin Immunoprecipitation Data. Bioinformatics 22: 423–429.

141. HochedlingerK, YamadaY, BeardC, JaenischR (2005) Ectopic Expression of Oct-4 Blocks Progenitor-Cell Differentiation and Causes Dysplasia in Epithelial Tissues. Cell 121: 465–477.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 2
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#