#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

An Regulatory Circuit Modulates /Wnt Signaling and Determines the Size of the Midbrain Dopaminergic Progenitor Pool


MicroRNAs regulate gene expression in diverse physiological scenarios. Their role in the control of morphogen related signaling pathways has been less studied, particularly in the context of embryonic Central Nervous System (CNS) development. Here, we uncover a role for microRNAs in limiting the spatiotemporal range of morphogen expression and function. Wnt1 is a key morphogen in the embryonic midbrain, and directs proliferation, survival, patterning and neurogenesis. We reveal an autoregulatory negative feedback loop between the transcription factor Lmx1b and a newly characterized microRNA, miR135a2, which modulates the extent of Wnt1/Wnt signaling and the size of the dopamine progenitor domain. Conditional gain of function studies reveal that Lmx1b promotes Wnt1/Wnt signaling, and thereby increases midbrain size and dopamine progenitor allocation. Conditional removal of Lmx1b has the opposite effect, in that expansion of the dopamine progenitor domain is severely compromised. Next, we provide evidence that microRNAs are involved in restricting dopamine progenitor allocation. Conditional loss of Dicer1 in embryonic stem cells (ESCs) results in expanded Lmx1a/b+ progenitors. In contrast, forced elevation of miR135a2 during an early window in vivo phenocopies the Lmx1b conditional knockout. When En1::Cre, but not Shh::Cre or Nes::Cre, is used for recombination, the expansion of Lmx1a/b+ progenitors is selectively reduced. Bioinformatics and luciferase assay data suggests that miR135a2 targets Lmx1b and many genes in the Wnt signaling pathway, including Ccnd1, Gsk3b, and Tcf7l2. Consistent with this, we demonstrate that this mutant displays reductions in the size of the Lmx1b/Wnt1 domain and range of canonical Wnt signaling. We posit that microRNA modulation of the Lmx1b/Wnt axis in the early midbrain/isthmus could determine midbrain size and allocation of dopamine progenitors. Since canonical Wnt activity has recently been recognized as a key ingredient for programming ESCs towards a dopaminergic fate in vitro, these studies could impact the rational design of such protocols.


Vyšlo v časopise: An Regulatory Circuit Modulates /Wnt Signaling and Determines the Size of the Midbrain Dopaminergic Progenitor Pool. PLoS Genet 9(12): e32767. doi:10.1371/journal.pgen.1003973
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003973

Souhrn

MicroRNAs regulate gene expression in diverse physiological scenarios. Their role in the control of morphogen related signaling pathways has been less studied, particularly in the context of embryonic Central Nervous System (CNS) development. Here, we uncover a role for microRNAs in limiting the spatiotemporal range of morphogen expression and function. Wnt1 is a key morphogen in the embryonic midbrain, and directs proliferation, survival, patterning and neurogenesis. We reveal an autoregulatory negative feedback loop between the transcription factor Lmx1b and a newly characterized microRNA, miR135a2, which modulates the extent of Wnt1/Wnt signaling and the size of the dopamine progenitor domain. Conditional gain of function studies reveal that Lmx1b promotes Wnt1/Wnt signaling, and thereby increases midbrain size and dopamine progenitor allocation. Conditional removal of Lmx1b has the opposite effect, in that expansion of the dopamine progenitor domain is severely compromised. Next, we provide evidence that microRNAs are involved in restricting dopamine progenitor allocation. Conditional loss of Dicer1 in embryonic stem cells (ESCs) results in expanded Lmx1a/b+ progenitors. In contrast, forced elevation of miR135a2 during an early window in vivo phenocopies the Lmx1b conditional knockout. When En1::Cre, but not Shh::Cre or Nes::Cre, is used for recombination, the expansion of Lmx1a/b+ progenitors is selectively reduced. Bioinformatics and luciferase assay data suggests that miR135a2 targets Lmx1b and many genes in the Wnt signaling pathway, including Ccnd1, Gsk3b, and Tcf7l2. Consistent with this, we demonstrate that this mutant displays reductions in the size of the Lmx1b/Wnt1 domain and range of canonical Wnt signaling. We posit that microRNA modulation of the Lmx1b/Wnt axis in the early midbrain/isthmus could determine midbrain size and allocation of dopamine progenitors. Since canonical Wnt activity has recently been recognized as a key ingredient for programming ESCs towards a dopaminergic fate in vitro, these studies could impact the rational design of such protocols.


Zdroje

1. KhudayberdievS, FioreR, SchrattG (2009) MicroRNA as modulators of neuronal responses. Commun Integr Biol 2: 411–413.

2. FinebergSK, KosikKS, DavidsonBL (2009) MicroRNAs potentiate neural development. Neuron 64: 303–309.

3. HeX, YuY, AwatramaniR, LuQR (2012) Unwrapping myelination by microRNAs. Neuroscientist 18: 45–55.

4. YunB, AndereggA, MenichellaD, WrabetzL, FeltriML, et al. (2010) MicroRNA-deficient Schwann cells display congenital hypomyelination. J Neurosci 30: 7722–7728.

5. ChenJA, HuangYP, MazzoniEO, TanGC, ZavadilJ, et al. (2011) Mir-17-3p controls spinal neural progenitor patterning by regulating Olig2/Irx3 cross-repressive loop. Neuron 69: 721–735.

6. Shi Y, Zhao X, Hsieh J, Wichterle H, Impey S, et al. MicroRNA regulation of neural stem cells and neurogenesis. J Neurosci 30: 14931–14936.

7. PengC, LiN, NgYK, ZhangJ, MeierF, et al. (2012) A unilateral negative feedback loop between miR-200 microRNAs and Sox2/E2F3 controls neural progenitor cell-cycle exit and differentiation. J Neurosci 32: 13292–13308.

8. HarfeBD, ScherzPJ, NissimS, TianH, McMahonAP, et al. (2004) Evidence for an expansion-based temporal Shh gradient in specifying vertebrate digit identities. Cell 118: 517–528.

9. DessaudE, YangLL, HillK, CoxB, UlloaF, et al. (2007) Interpretation of the sonic hedgehog morphogen gradient by a temporal adaptation mechanism. Nature 450: 717–720.

10. AhnS, JoynerAL (2004) Dynamic changes in the response of cells to positive hedgehog signaling during mouse limb patterning. Cell 118: 505–516.

11. MartelloG, ZacchignaL, InuiM, MontagnerM, AdornoM, et al. (2007) MicroRNA control of Nodal signalling. Nature 449: 183–188.

12. InestrosaNC, ArenasE (2010) Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci 11: 77–86.

13. Alves dos SantosMT, SmidtMP (2011) En1 and Wnt signaling in midbrain dopaminergic neuronal development. Neural Dev 6: 23.

14. UlloaF, BriscoeJ (2007) Morphogens and the control of cell proliferation and patterning in the spinal cord. Cell Cycle 6: 2640–2649.

15. AnderssonER, SaltoC, VillaescusaJC, CajanekL, YangS, et al. (2013) Wnt5a cooperates with canonical Wnts to generate midbrain dopaminergic neurons in vivo and in stem cells. Proc Natl Acad Sci U S A 110: E602–610.

16. YangJ, BrownA, EllisorD, PaulE, HaganN, et al. (2013) Dynamic temporal requirement of Wnt1 in midbrain dopamine neuron development. Development 140: 1342–1352.

17. JoksimovicM, YunBA, KittappaR, AndereggAM, ChangWW, et al. (2009) Wnt antagonism of Shh facilitates midbrain floor plate neurogenesis. Nat Neurosci 12: 125–131.

18. ChilovD, SinjushinaN, Saarimaki-VireJ, TaketoMM, PartanenJ (2010) beta-Catenin regulates intercellular signalling networks and cell-type specific transcription in the developing mouse midbrain-rhombomere 1 region. PLoS ONE 5: e10881.

19. TangM, MiyamotoY, HuangEJ (2009) Multiple roles of beta-catenin in controlling the neurogenic niche for midbrain dopamine neurons. Development 136: 2027–2038.

20. TangM, VillaescusaJC, LuoSX, GuitarteC, LeiS, et al. (2010) Interactions of Wnt/beta-catenin signaling and sonic hedgehog regulate the neurogenesis of ventral midbrain dopamine neurons. J Neurosci 30: 9280–9291.

21. CajanekL, RibeiroD, ListeI, ParishCL, BryjaV, et al. (2009) Wnt/beta-catenin signaling blockade promotes neuronal induction and dopaminergic differentiation in embryonic stem cells. Stem Cells 27: 2917–2927.

22. JoksimovicM, AndereggA, RoyA, CampochiaroL, YunB, et al. (2009) Spatiotemporally separable Shh domains in the midbrain define distinct dopaminergic progenitor pools. Proc Natl Acad Sci U S A 106: 19185–19190.

23. Blaess S, Bodea GO, Kabanova A, Chanet S, Mugniery E, et al. Temporal-spatial changes in Sonic Hedgehog expression and signaling reveal different potentials of ventral mesencephalic progenitors to populate distinct ventral midbrain nuclei. Neural Dev 6: 29.

24. AgarwalaS, RagsdaleCW (2002) A role for midbrain arcs in nucleogenesis. Development 129: 5779–5788.

25. NakataniT, MinakiY, KumaiM, OnoY (2007) Helt determines GABAergic over glutamatergic neuronal fate by repressing Ngn genes in the developing mesencephalon. Development 134: 2783–2793.

26. PrakashN, PuellesE, FreudeK, TrumbachD, OmodeiD, et al. (2009) Nkx6-1 controls the identity and fate of red nucleus and oculomotor neurons in the mouse midbrain. Development 136: 2545–2555.

27. AnderssonE, TryggvasonU, DengQ, FrilingS, AlekseenkoZ, et al. (2006) Identification of intrinsic determinants of midbrain dopamine neurons. Cell 124: 393–405.

28. BlaessS, BodeaGO, KabanovaA, ChanetS, MugnieryE, et al. (2011) Temporal-spatial changes in Sonic Hedgehog expression and signaling reveal different potentials of ventral mesencephalic progenitors to populate distinct ventral midbrain nuclei. Neural Dev 6: 29.

29. PrakashN, BrodskiC, NaserkeT, PuellesE, GogoiR, et al. (2006) A Wnt1-regulated genetic network controls the identity and fate of midbrain-dopaminergic progenitors in vivo. Development 133: 89–98.

30. TangM, MiyamotoY, HuangEJ (2009) Multiple roles of {beta}-catenin in controlling the neurogenic niche for midbrain dopamine neurons. Development 136: 2027–2038.

31. Castelo-BrancoG, WagnerJ, RodriguezFJ, KeleJ, SousaK, et al. (2003) Differential regulation of midbrain dopaminergic neuron development by Wnt-1, Wnt-3a, and Wnt-5a. Proc Natl Acad Sci U S A 100: 12747–12752.

32. JoksimovicM, PatelM, TaketoMM, JohnsonR, AwatramaniR (2012) Ectopic wnt/beta-catenin signaling induces neurogenesis in the spinal cord and hindbrain floor plate. PLoS ONE 7: e30266.

33. LewisBP, BurgeCB, BartelDP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120: 15–20.

34. MansfieldJH, HarfeBD, NissenR, ObenauerJ, SrineelJ, et al. (2004) MicroRNA-responsive ‘sensor’ transgenes uncover Hox-like and other developmentally regulated patterns of vertebrate microRNA expression. Nat Genet 36: 1079–1083.

35. RockJR, LopezMC, BakerHV, HarfeBD (2007) Identification of genes expressed in the mouse limb using a novel ZPA microarray approach. Gene Expr Patterns 8: 19–26.

36. UhdeCW, VivesJ, JaegerI, LiM (2010) Rmst is a novel marker for the mouse ventral mesencephalic floor plate and the anterior dorsal midline cells. PLoS ONE 5: e8641.

37. GromakN (2012) Intronic microRNAs: a crossroad in gene regulation. Biochem Soc Trans 40: 759–761.

38. GuoC, QiuHY, HuangY, ChenH, YangRQ, et al. (2007) Lmx1b is essential for Fgf8 and Wnt1 expression in the isthmic organizer during tectum and cerebellum development in mice. Development 134: 317–325.

39. AdamsKA, MaidaJM, GoldenJA, RiddleRD (2000) The transcription factor Lmx1b maintains Wnt1 expression within the isthmic organizer. Development 127: 1857–1867.

40. StarkA, BrenneckeJ, BushatiN, RussellRB, CohenSM (2005) Animal MicroRNAs confer robustness to gene expression and have a significant impact on 3′UTR evolution. Cell 123: 1133–1146.

41. KimmelRA, TurnbullDH, BlanquetV, WurstW, LoomisCA, et al. (2000) Two lineage boundaries coordinate vertebrate apical ectodermal ridge formation. Genes Dev 14: 1377–1389.

42. LiY, QiuQ, WatsonSS, SchweitzerR, JohnsonRL (2010) Uncoupling skeletal and connective tissue patterning: conditional deletion in cartilage progenitors reveals cell-autonomous requirements for Lmx1b in dorsal-ventral limb patterning. Development 137: 1181–1188.

43. JhoEH, ZhangT, DomonC, JooCK, FreundJN, et al. (2002) Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol 22: 1172–1183.

44. ZengYA, NusseR (2010) Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture. Cell Stem Cell 6: 568–577.

45. ZhaoZQ, ScottM, ChiechioS, WangJS, RennerKJ, et al. (2006) Lmx1b is required for maintenance of central serotonergic neurons and mice lacking central serotonergic system exhibit normal locomotor activity. J Neurosci 26: 12781–12788.

46. ChungS, LeungA, HanBS, ChangMY, MoonJI, et al. (2009) Wnt1-lmx1a forms a novel autoregulatory loop and controls midbrain dopaminergic differentiation synergistically with the SHH-FoxA2 pathway. Cell Stem Cell 5: 646–658.

47. LinW, MetzakopianE, MavromatakisYE, GaoN, BalaskasN, et al. (2009) Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development. Dev Biol 333: 386–396.

48. NakataniT, KumaiM, MizuharaE, MinakiY, OnoY (2010) Lmx1a and Lmx1b cooperate with Foxa2 to coordinate the specification of dopaminergic neurons and control of floor plate cell differentiation in the developing mesencephalon. Dev Biol 339: 101–113.

49. WichterleH, LieberamI, PorterJA, JessellTM (2002) Directed differentiation of embryonic stem cells into motor neurons. Cell 110: 385–397.

50. DengQ, AnderssonE, HedlundE, AlekseenkoZ, CoppolaE, et al. (2011) Specific and integrated roles of Lmx1a, Lmx1b and Phox2a in ventral midbrain development. Development 138: 3399–3408.

51. YanCH, LevesqueM, ClaxtonS, JohnsonRL, AngSL (2011) Lmx1a and lmx1b function cooperatively to regulate proliferation, specification, and differentiation of midbrain dopaminergic progenitors. J Neurosci 31: 12413–12425.

52. SmidtMP, AsbreukCH, CoxJJ, ChenH, JohnsonRL, et al. (2000) A second independent pathway for development of mesencephalic dopaminergic neurons requires Lmx1b. Nat Neurosci 3: 337–341.

53. DavisCA, JoynerAL (1988) Expression patterns of the homeo box-containing genes En-1 and En-2 and the proto-oncogene int-1 diverge during mouse development. Genes Dev 2: 1736–1744.

54. ChiCL, MartinezS, WurstW, MartinGR (2003) The isthmic organizer signal FGF8 is required for cell survival in the prospective midbrain and cerebellum. Development 130: 2633–2644.

55. EchelardY, EpsteinDJ, St-JacquesB, ShenL, MohlerJ, et al. (1993) Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity. Cell 75: 1417–1430.

56. VernayB, KochM, VaccarinoF, BriscoeJ, SimeoneA, et al. (2005) Otx2 regulates subtype specification and neurogenesis in the midbrain. J Neurosci 25: 4856–4867.

57. LiuA, LososK, JoynerAL (1999) FGF8 can activate Gbx2 and transform regions of the rostral mouse brain into a hindbrain fate. Development 126: 4827–4838.

58. HornsteinE, ShomronN (2006) Canalization of development by microRNAs. Nat Genet 38 Suppl: S20–24.

59. InestrosaNC, ArenasE (2010) Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci 11: 77–86.

60. PanhuysenM, Vogt WeisenhornDM, BlanquetV, BrodskiC, HeinzmannU, et al. (2004) Effects of Wnt1 signaling on proliferation in the developing mid-/hindbrain region. Mol Cell Neurosci 26: 101–111.

61. MatsunagaE, KatahiraT, NakamuraH (2002) Role of Lmx1b and Wnt1 in mesencephalon and metencephalon development. Development 129: 5269–5277.

62. McMahonAP, BradleyA (1990) The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell 62: 1073–1085.

63. OmodeiD, AcamporaD, MancusoP, PrakashN, Di GiovannantonioLG, et al. (2008) Anterior-posterior graded response to Otx2 controls proliferation and differentiation of dopaminergic progenitors in the ventral mesencephalon. Development 135: 3459–3470.

64. CaiJ, SchleidtS, Pelta-HellerJ, HutchingsD, CannarsaG, et al. (2013) BMP and TGF-beta pathway mediators are critical upstream regulators of Wnt signaling during midbrain dopamine differentiation in human pluripotent stem cells. Dev Biol 376: 62–73.

65. FalkS, WurdakH, IttnerLM, IlleF, SumaraG, et al. (2008) Brain area-specific effect of TGF-beta signaling on Wnt-dependent neural stem cell expansion. Cell Stem Cell 2: 472–483.

66. Perez-BalaguerA, PuellesE, WurstW, MartinezS (2009) Shh dependent and independent maintenance of basal midbrain. Mech Dev 126: 301–313.

67. Saarimaki-VireJ, PeltopuroP, LahtiL, NaserkeT, BlakAA, et al. (2007) Fibroblast growth factor receptors cooperate to regulate neural progenitor properties in the developing midbrain and hindbrain. J Neurosci 27: 8581–8592.

68. NagelR, le SageC, DiosdadoB, van der WaalM, Oude VrielinkJA, et al. (2008) Regulation of the adenomatous polyposis coli gene by the miR-135 family in colorectal cancer. Cancer Res 68: 5795–5802.

69. MendellJT, OlsonEN (2012) MicroRNAs in stress signaling and human disease. Cell 148: 1172–1187.

70. KriksS, ShimJW, PiaoJ, GanatYM, WakemanDR, et al. (2011) Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature 480: 547–51.

71. KirkebyA, GrealishS, WolfDA, NelanderJ, WoodJ, et al. (2012) Generation of regionally specified neural progenitors and functional neurons from human embryonic stem cells under defined conditions. Cell Rep 1: 703–714.

72. Anokye-DansoF, TrivediCM, JuhrD, GuptaM, CuiZ, et al. (2011) Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8: 376–388.

73. TroncheF, KellendonkC, KretzO, GassP, AnlagK, et al. (1999) Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat Genet 23: 99–103.

74. MacdonaldBT, SemenovMV, HeX (2007) SnapShot: Wnt/beta-catenin signaling. Cell 131: 1204.

75. TanakaSS, KojimaY, YamaguchiYL, NishinakamuraR, TamPP (2011) Impact of WNT signaling on tissue lineage differentiation in the early mouse embryo. Dev Growth Differ 53: 843–856.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 12
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#