#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Yap- and Cdc42-Dependent Nephrogenesis and Morphogenesis during Mouse Kidney Development


Yap is a transcriptional co-activator that regulates cell proliferation and apoptosis downstream of the Hippo kinase pathway. We investigated Yap function during mouse kidney development using a conditional knockout strategy that specifically inactivated Yap within the nephrogenic lineage. We found that Yap is essential for nephron induction and morphogenesis, surprisingly, in a manner independent of regulation of cell proliferation and apoptosis. We used microarray analysis to identify a suite of novel Yap-dependent genes that function during nephron formation and have been implicated in morphogenesis. Previous in vitro studies have indicated that Yap can respond to mechanical stresses in cultured cells downstream of the small GTPases RhoA. We find that tissue-specific inactivation of the Rho GTPase Cdc42 causes a severe defect in nephrogenesis that strikingly phenocopies loss of Yap. Ablation of Cdc42 decreases nuclear localization of Yap, leading to a reduction of Yap-dependent gene expression. We propose that Yap responds to Cdc42-dependent signals in nephron progenitor cells to activate a genetic program required to shape the functioning nephron.


Vyšlo v časopise: Yap- and Cdc42-Dependent Nephrogenesis and Morphogenesis during Mouse Kidney Development. PLoS Genet 9(3): e32767. doi:10.1371/journal.pgen.1003380
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003380

Souhrn

Yap is a transcriptional co-activator that regulates cell proliferation and apoptosis downstream of the Hippo kinase pathway. We investigated Yap function during mouse kidney development using a conditional knockout strategy that specifically inactivated Yap within the nephrogenic lineage. We found that Yap is essential for nephron induction and morphogenesis, surprisingly, in a manner independent of regulation of cell proliferation and apoptosis. We used microarray analysis to identify a suite of novel Yap-dependent genes that function during nephron formation and have been implicated in morphogenesis. Previous in vitro studies have indicated that Yap can respond to mechanical stresses in cultured cells downstream of the small GTPases RhoA. We find that tissue-specific inactivation of the Rho GTPase Cdc42 causes a severe defect in nephrogenesis that strikingly phenocopies loss of Yap. Ablation of Cdc42 decreases nuclear localization of Yap, leading to a reduction of Yap-dependent gene expression. We propose that Yap responds to Cdc42-dependent signals in nephron progenitor cells to activate a genetic program required to shape the functioning nephron.


Zdroje

1. NyengaardJR, BendtsenTF (1992) Glomerular number and size in relation to age, kidney weight, and body surface in normal man. Anat Rec 232: 194–201.

2. KellerG, ZimmerG, MallG, RitzE, AmannK (2003) Nephron number in patients with primary hypertension. N Engl J Med 348: 101–108.

3. SaxenL, SariolaH, LehtonenE (1986) Sequential cell and tissue interactions governing organogenesis of the kidney. Anat Embryol (Berl) 175: 1–6.

4. LittleMH, McMahonAP (2012) Mammalian kidney development: principles, progress, and projections. Cold Spring Harb Perspect Biol 4(5): pii: a008300 doi:10.1101/cshperspect.a008300

5. CostantiniF, KopanR Patterning a complex organ: branching morphogenesis and nephron segmentation in kidney development. Dev Cell 18: 698–712.

6. DresslerGR (2006) The cellular basis of kidney development. Annu Rev Cell Dev Biol 22: 509–529.

7. StaleyBK, IrvineKD Hippo signaling in Drosophila: recent advances and insights. Dev Dyn 241: 3–15.

8. ZhaoB, LeiQY, GuanKL (2008) The Hippo-YAP pathway: new connections between regulation of organ size and cancer. Curr Opin Cell Biol 20: 638–646.

9. HalderG, JohnsonRL Hippo signaling: growth control and beyond. Development 138: 9–22.

10. PanD The hippo signaling pathway in development and cancer. Dev Cell 19: 491–505.

11. HarveyK, TaponN (2007) The Salvador-Warts-Hippo pathway - an emerging tumour-suppressor network. Nat Rev Cancer 7: 182–191.

12. Morin-KensickiEM, BooneBN, HowellM, StonebrakerJR, TeedJ, et al. (2006) Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol Cell Biol 26: 77–87.

13. HossainZ, AliSM, KoHL, XuJ, NgCP, et al. (2007) Glomerulocystic kidney disease in mice with a targeted inactivation of Wwtr1. Proc Natl Acad Sci U S A 104: 1631–1636.

14. MakitaR, UchijimaY, NishiyamaK, AmanoT, ChenQ, et al. (2008) Multiple renal cysts, urinary concentration defects, and pulmonary emphysematous changes in mice lacking TAZ. Am J Physiol Renal Physiol 294: F542–553.

15. NishiokaN, InoueK, AdachiK, KiyonariH, OtaM, et al. (2009) The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell 16: 398–410.

16. CamargoFD, GokhaleS, JohnnidisJB, FuD, BellGW, et al. (2007) YAP1 increases organ size and expands undifferentiated progenitor cells. Curr Biol 17: 2054–2060.

17. LeeJH, KimTS, YangTH, KooBK, OhSP, et al. (2008) A crucial role of WW45 in developing epithelial tissues in the mouse. Embo J 27: 1231–1242.

18. HeallenT, ZhangM, WangJ, Bonilla-ClaudioM, KlysikE, et al. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 332: 458–461.

19. YuFX, ZhaoB, PanupinthuN, JewellJL, LianI, et al. Regulation of the Hippo-YAP Pathway by G-Protein-Coupled Receptor Signaling. Cell 150: 780–791.

20. BoggianoJC, FehonRG Growth control by committee: intercellular junctions, cell polarity, and the cytoskeleton regulate Hippo signaling. Dev Cell 22: 695–702.

21. DupontS, MorsutL, AragonaM, EnzoE, GiulittiS, et al. Role of YAP/TAZ in mechanotransduction. Nature 474: 179–183.

22. WadaK, ItogaK, OkanoT, YonemuraS, SasakiH Hippo pathway regulation by cell morphology and stress fibers. Development 138: 3907–3914.

23. ZhaoB, TumanengK, GuanKL The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat Cell Biol 13: 877–883.

24. DongJ, FeldmannG, HuangJ, WuS, ZhangN, et al. (2007) Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 130: 1120–1133.

25. KobayashiA, ValeriusMT, MugfordJW, CarrollTJ, SelfM, et al. (2008) Six2 defines and regulates a multipotent self-renewing nephron progenitor population throughout mammalian kidney development. Cell Stem Cell 3: 169–181.

26. ReginensiA, ClarksonM, NeirijnckY, LuB, OhyamaT, et al. SOX9 controls epithelial branching by activating RET effector genes during kidney development. Hum Mol Genet 20: 1143–1153.

27. ChengHT, KimM, ValeriusMT, SurendranK, Schuster-GosslerK, et al. (2007) Notch2, but not Notch1, is required for proximal fate acquisition in the mammalian nephron. Development 134: 801–811.

28. ArmstrongJF, Pritchard-JonesK, BickmoreWA, HastieND, BardJB (1993) The expression of the Wilms' tumour gene, WT1, in the developing mammalian embryo. Mech Dev 40: 85–97.

29. GeorgasK, RumballeB, ValeriusMT, ChiuHS, ThiagarajanRD, et al. (2009) Analysis of early nephron patterning reveals a role for distal RV proliferation in fusion to the ureteric tip via a cap mesenchyme-derived connecting segment. Dev Biol 332: 273–286.

30. VarelasX, MillerBW, SopkoR, SongS, GregorieffA, et al. The Hippo pathway regulates Wnt/beta-catenin signaling. Dev Cell 18: 579–591.

31. ParkJS, ValeriusMT, McMahonAP (2007) Wnt/beta-catenin signaling regulates nephron induction during mouse kidney development. Development 134: 2533–2539.

32. CarrollTJ, ParkJS, HayashiS, MajumdarA, McMahonAP (2005) Wnt9b plays a central role in the regulation of mesenchymal to epithelial transitions underlying organogenesis of the mammalian urogenital system. Dev Cell 9: 283–292.

33. RohatgiR, MaL, MikiH, LopezM, KirchhausenT, et al. (1999) The interaction between N-WASP and the Arp2/3 complex links Cdc42-dependent signals to actin assembly. Cell 97: 221–231.

34. TonamiK, KuriharaY, AburataniH, UchijimaY, AsanoT, et al. (2007) Calpain 6 is involved in microtubule stabilization and cytoskeletal organization. Mol Cell Biol 27: 2548–2561.

35. LianI, KimJ, OkazawaH, ZhaoJ, ZhaoB, et al. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation. Genes Dev 24: 1106–1118.

36. TorresM, Gomez-PardoE, DresslerGR, GrussP (1995) Pax-2 controls multiple steps of urogenital development. Development 121: 4057–4065.

37. JaffeAB, HallA (2005) Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol 21: 247–269.

38. WuX, QuondamatteoF, LefeverT, CzuchraA, MeyerH, et al. (2006) Cdc42 controls progenitor cell differentiation and beta-catenin turnover in skin. Genes Dev 20: 571–585.

39. OhS, LeeD, KimT, KimTS, OhHJ, et al. (2009) Crucial role for Mst1 and Mst2 kinases in early embryonic development of the mouse. Mol Cell Biol 29: 6309–6320.

40. Cotta-de-AlmeidaV, WesterbergL, MaillardMH, OnaldiD, WachtelH, et al. (2007) Wiskott Aldrich syndrome protein (WASP) and N-WASP are critical for T cell development. Proc Natl Acad Sci U S A 104: 15424–15429.

41. EnglekaKA, GitlerAD, ZhangM, ZhouDD, HighFA, et al. (2005) Insertion of Cre into the Pax3 locus creates a new allele of Splotch and identifies unexpected Pax3 derivatives. Dev Biol 280: 396–406.

42. WilkinsonDG (1995) RNA detection using non-radioactive in situ hybridization. Curr Opin Biotechnol 6: 20–23.

43. RosselotC, SpraggonL, ChiaI, BatourinaE, RiccioP, et al. Non-cell-autonomous retinoid signaling is crucial for renal development. Development 137: 283–292.

44. MichosO, CebrianC, HyinkD, GrieshammerU, WilliamsL, et al. Kidney development in the absence of Gdnf and Spry1 requires Fgf10. PLoS Genet 6: e1000809 doi:10.1371/journal.pgen.1000809

45. OlhovskyM, WillitonK, DaiAY, PasculescuA, LeeJP, et al. OpenFreezer: a reagent information management software system. Nat Methods 8: 612–613.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 3
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#