#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Ubiquitin Ligase Subunit Acts in Target Tissue to Restrict Tracheal Terminal Cell Branching and Hypoxic-Induced Gene Expression


The Drosophila melanogaster gene archipelago (ago) encodes the F-box/WD-repeat protein substrate specificity factor for an SCF (Skp/Cullin/F-box)-type polyubiquitin ligase that inhibits tumor-like growth by targeting proteins for degradation by the proteasome. The Ago protein is expressed widely in the fly embryo and larva and promotes degradation of pro-proliferative proteins in mitotically active cells. However the requirement for Ago in post-mitotic developmental processes remains largely unexplored. Here we show that Ago is an antagonist of the physiologic response to low oxygen (hypoxia). Reducing Ago activity in larval muscle cells elicits enhanced branching of nearby tracheal terminal cells in normoxia. This tracheogenic phenotype shows a genetic dependence on sima, which encodes the HIF-1α subunit of the hypoxia-inducible transcription factor dHIF and its target the FGF ligand branchless (bnl), and is enhanced by depletion of the Drosophila Von Hippel Lindau (dVHL) factor, which is a subunit of an oxygen-dependent ubiquitin ligase that degrades Sima/HIF-1α protein in metazoan cells. Genetic reduction of ago results in constitutive expression of some hypoxia-inducible genes in normoxia, increases the sensitivity of others to mild hypoxic stimulus, and enhances the ability of adult flies to recover from hypoxic stupor. As a molecular correlate to these genetic data, we find that Ago physically associates with Sima and restricts Sima levels in vivo. Collectively, these findings identify Ago as a required element of a circuit that suppresses the tracheogenic activity of larval muscle cells by antagonizing the Sima-mediated transcriptional response to hypoxia.


Vyšlo v časopise: The Ubiquitin Ligase Subunit Acts in Target Tissue to Restrict Tracheal Terminal Cell Branching and Hypoxic-Induced Gene Expression. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003314
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003314

Souhrn

The Drosophila melanogaster gene archipelago (ago) encodes the F-box/WD-repeat protein substrate specificity factor for an SCF (Skp/Cullin/F-box)-type polyubiquitin ligase that inhibits tumor-like growth by targeting proteins for degradation by the proteasome. The Ago protein is expressed widely in the fly embryo and larva and promotes degradation of pro-proliferative proteins in mitotically active cells. However the requirement for Ago in post-mitotic developmental processes remains largely unexplored. Here we show that Ago is an antagonist of the physiologic response to low oxygen (hypoxia). Reducing Ago activity in larval muscle cells elicits enhanced branching of nearby tracheal terminal cells in normoxia. This tracheogenic phenotype shows a genetic dependence on sima, which encodes the HIF-1α subunit of the hypoxia-inducible transcription factor dHIF and its target the FGF ligand branchless (bnl), and is enhanced by depletion of the Drosophila Von Hippel Lindau (dVHL) factor, which is a subunit of an oxygen-dependent ubiquitin ligase that degrades Sima/HIF-1α protein in metazoan cells. Genetic reduction of ago results in constitutive expression of some hypoxia-inducible genes in normoxia, increases the sensitivity of others to mild hypoxic stimulus, and enhances the ability of adult flies to recover from hypoxic stupor. As a molecular correlate to these genetic data, we find that Ago physically associates with Sima and restricts Sima levels in vivo. Collectively, these findings identify Ago as a required element of a circuit that suppresses the tracheogenic activity of larval muscle cells by antagonizing the Sima-mediated transcriptional response to hypoxia.


Zdroje

1. DenkoNC (2008) Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat Rev Cancer 8: 705–713.

2. KaelinWGJr, RatcliffePJ (2008) Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell 30: 393–402.

3. GorrTA, GassmannM, WappnerP (2006) Sensing and responding to hypoxia via HIF in model invertebrates. J Insect Physiol 52: 349–364.

4. WangGL, JiangBH, RueEA, SemenzaGL (1995) Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A 92: 5510–5514.

5. WangGL, JiangBH, SemenzaGL (1995) Effect of altered redox states on expression and DNA-binding activity of hypoxia-inducible factor 1. Biochem Biophys Res Commun 212: 550–556.

6. WangGL, SemenzaGL (1995) Purification and characterization of hypoxia-inducible factor 1. J Biol Chem 270: 1230–1237.

7. SemenzaGL, WangGL (1992) A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12: 5447–5454.

8. BaconNC, WappnerP, O'RourkeJF, BartlettSM, ShiloB, et al. (1998) Regulation of the Drosophila bHLH-PAS protein Sima by hypoxia: functional evidence for homology with mammalian HIF-1 alpha. Biochem Biophys Res Commun 249: 811–816.

9. NambuJR, ChenW, HuS, CrewsST (1996) The Drosophila melanogaster similar bHLH-PAS gene encodes a protein related to human hypoxia-inducible factor 1 alpha and Drosophila single-minded. Gene 172: 249–254.

10. SonnenfeldM, WardM, NystromG, MosherJ, StahlS, et al. (1997) The Drosophila tango gene encodes a bHLH-PAS protein that is orthologous to mammalian Arnt and controls CNS midline and tracheal development. Development 124: 4571–4582.

11. OhshiroT, SaigoK (1997) Transcriptional regulation of breathless FGF receptor gene by binding of TRACHEALESS/dARNT heterodimers to three central midline elements in Drosophila developing trachea. Development 124: 3975–3986.

12. MaE, HaddadGG (1999) Isolation and characterization of the hypoxia-inducible factor 1beta in Drosophila melanogaster. Brain Res Mol Brain Res 73: 11–16.

13. WeidemannA, JohnsonRS (2008) Biology of HIF-1alpha. Cell Death Differ 15: 621–627.

14. BruickRK, McKnightSL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294: 1337–1340.

15. EpsteinAC, GleadleJM, McNeillLA, HewitsonKS, O'RourkeJ, et al. (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107: 43–54.

16. IvanM, KondoK, YangH, KimW, ValiandoJ, et al. (2001) HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292: 464–468.

17. JaakkolaP, MoleDR, TianYM, WilsonMI, GielbertJ, et al. (2001) Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292: 468–472.

18. CockmanME, MassonN, MoleDR, JaakkolaP, ChangGW, et al. (2000) Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. J Biol Chem 275: 25733–25741.

19. OhhM, ParkCW, IvanM, HoffmanMA, KimTY, et al. (2000) Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2: 423–427.

20. MaxwellPH, WiesenerMS, ChangGW, CliffordSC, VauxEC, et al. (1999) The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399: 271–275.

21. ArquierN, VigneP, DuplanE, HsuT, TherondPP, et al. (2006) Analysis of the hypoxia-sensing pathway in Drosophila melanogaster. Biochem J 393: 471–480.

22. CentaninL, DekantyA, RomeroN, IrisarriM, GorrTA, et al. (2008) Cell autonomy of HIF effects in Drosophila: tracheal cells sense hypoxia and induce terminal branch sprouting. Dev Cell 14: 547–558.

23. CentaninL, RatcliffePJ, WappnerP (2005) Reversion of lethality and growth defects in Fatiga oxygen-sensor mutant flies by loss of Hypoxia-Inducible Factor-alpha/Sima. EMBO Rep 6: 1070–1075.

24. Lavista-LlanosS, CentaninL, IrisarriM, RussoDM, GleadleJM, et al. (2002) Control of the hypoxic response in Drosophila melanogaster by the basic helix-loop-helix PAS protein similar. Mol Cell Biol 22: 6842–6853.

25. KondoK, KaelinWGJr (2001) The von Hippel-Lindau tumor suppressor gene. Exp Cell Res 264: 117–125.

26. WarburgO (1956) On respiratory impairment in cancer cells. Science 124: 269–270.

27. RankinEB, GiacciaAJ (2008) The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ 15: 678–685.

28. ZhouJ, SchmidT, SchnitzerS, BruneB (2006) Tumor hypoxia and cancer progression. Cancer Lett 237: 10–21.

29. RomeroNM, DekantyA, WappnerP (2007) Cellular and developmental adaptations to hypoxia: a Drosophila perspective. Methods Enzymol 435: 123–144.

30. HoogewijsD, GeuensE, DewildeS, VierstraeteA, MoensL, et al. (2007) Wide diversity in structure and expression profiles among members of the Caenorhabditis elegans globin protein family. BMC Genomics 8: 356.

31. LiuG, RoyJ, JohnsonEA (2006) Identification and function of hypoxia-response genes in Drosophila melanogaster. Physiol Genomics 25: 134–141.

32. ZhouD, XueJ, LaiJC, SchorkNJ, WhiteKP, et al. (2008) Mechanisms underlying hypoxia tolerance in Drosophila melanogaster: hairy as a metabolic switch. PLoS Genet 4: e1000221 doi:10.1371/journal.pgen.1000221

33. HaddadGG, SunY, WymanRJ, XuT (1997) Genetic basis of tolerance to O2 deprivation in Drosophila melanogaster. Proc Natl Acad Sci U S A 94: 10809–10812.

34. HaddadGG, WymanRJ, MohseninA, SunY, KrishnanSN (1997) Behavioral and Electrophysiologic Responses of Drosophila melanogaster to Prolonged Periods of Anoxia. J Insect Physiol 43: 203–210.

35. MaE, HaddadGG (1997) Anoxia regulates gene expression in the central nervous system of Drosophila melanogaster. Brain Res Mol Brain Res 46: 325–328.

36. MaE, XuT, HaddadGG (1999) Gene regulation by O2 deprivation: an anoxia-regulated novel gene in Drosophila melanogaster. Brain Res Mol Brain Res 63: 217–224.

37. CentaninL, GorrTA, WappnerP (2010) Tracheal remodelling in response to hypoxia. J Insect Physiol 56: 447–454.

38. JareckiJ, JohnsonE, KrasnowMA (1999) Oxygen regulation of airway branching in Drosophila is mediated by branchless FGF. Cell 99: 211–220.

39. KlambtC, GlazerL, ShiloBZ (1992) breathless, a Drosophila FGF receptor homolog, is essential for migration of tracheal and specific midline glial cells. Genes Dev 6: 1668–1678.

40. SutherlandD, SamakovlisC, KrasnowMA (1996) branchless encodes a Drosophila FGF homolog that controls tracheal cell migration and the pattern of branching. Cell 87: 1091–1101.

41. GuilleminK, GroppeJ, DuckerK, TreismanR, HafenE, et al. (1996) The pruned gene encodes the Drosophila serum response factor and regulates cytoplasmic outgrowth during terminal branching of the tracheal system. Development 122: 1353–1362.

42. SamakovlisC, ManningG, StenebergP, HacohenN, CanteraR, et al. (1996) Genetic control of epithelial tube fusion during Drosophila tracheal development. Development 122: 3531–3536.

43. FlugelD, GorlachA, MichielsC, KietzmannT (2007) Glycogen synthase kinase 3 phosphorylates hypoxia-inducible factor 1alpha and mediates its destabilization in a VHL-independent manner. Mol Cell Biol 27: 3253–3265.

44. ZhangD, LiJ, CostaM, GaoJ, HuangC (2010) JNK1 mediates degradation HIF-1alpha by a VHL-independent mechanism that involves the chaperones Hsp90/Hsp70. Cancer Res 70: 813–823.

45. FlugelD, GorlachA, KietzmannT (2012) GSK-3beta regulates cell growth, migration, and angiogenesis via Fbw7 and USP28-dependent degradation of HIF-1alpha. Blood 119: 1292–1301.

46. CassavaughJM, HaleSA, WellmanTL, HoweAK, WongC, et al. (2011) Negative regulation of HIF-1alpha by an FBW7-mediated degradation pathway during hypoxia. J Cell Biochem 112: 3882–3890.

47. MortimerNT, MobergKH (2007) The Drosophila F-box protein Archipelago controls levels of the Trachealess transcription factor in the embryonic tracheal system. Dev Biol 312: 560–571.

48. MortimerNT, MobergKH (2009) Regulation of Drosophila embryonic tracheogenesis by dVHL and hypoxia. Dev Biol 329: 294–305.

49. MobergKH, BellDW, WahrerDC, HaberDA, HariharanIK (2001) Archipelago regulates Cyclin E levels in Drosophila and is mutated in human cancer cell lines. Nature 413: 311–316.

50. MobergKH, MukherjeeA, VeraksaA, Artavanis-TsakonasS, HariharanIK (2004) The Drosophila F box protein archipelago regulates dMyc protein levels in vivo. Curr Biol 14: 965–974.

51. WelckerM, ClurmanBE (2008) FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer 8: 83–93.

52. GhabrialA, LuschnigS, MetzsteinMM, KrasnowMA (2003) Branching morphogenesis of the Drosophila tracheal system. Annu Rev Cell Dev Biol 19: 623–647.

53. AdryanB, DeckerHJ, PapasTS, HsuT (2000) Tracheal development and the von Hippel-Lindau tumor suppressor homolog in Drosophila. Oncogene 19: 2803–2811.

54. AsoT, YamazakiK, AigakiT, KitajimaS (2000) Drosophila von Hippel-Lindau tumor suppressor complex possesses E3 ubiquitin ligase activity. Biochem Biophys Res Commun 276: 355–361.

55. SemenzaGL (2007) Hypoxia-inducible factor 1 (HIF-1) pathway. Sci STKE 2007: cm8.

56. SemenzaGL (2007) Life with oxygen. Science 318: 62–64.

57. ErlerJT, BennewithKL, NicolauM, DornhoferN, KongC, et al. (2006) Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 440: 1222–1226.

58. ErlerJT, GiacciaAJ (2006) Lysyl oxidase mediates hypoxic control of metastasis. Cancer Res 66: 10238–10241.

59. RodriguezC, AlcudiaJF, Martinez-GonzalezJ, RaposoB, NavarroMA, et al. (2008) Lysyl oxidase (LOX) down-regulation by TNFalpha: a new mechanism underlying TNFalpha-induced endothelial dysfunction. Atherosclerosis 196: 558–564.

60. WangJ, CaoY, ChenY, GardnerP, SteinerDF (2006) Pancreatic beta cells lack a low glucose and O2-inducible mitochondrial protein that augments cell survival. Proc Natl Acad Sci U S A 103: 10636–10641.

61. AzadP, HaddadGG (2009) Survival in acute and severe low o environment: use of a genetic model system. Ann N Y Acad Sci 1177: 39–47.

62. JiangBH, SemenzaGL, BauerC, MartiHH (1996) Hypoxia-inducible factor 1 levels vary exponentially over a physiologically relevant range of O2 tension. Am J Physiol 271: C1172–1180.

63. SundqvistA, Bengoechea-AlonsoMT, YeX, LukiyanchukV, JinJ, et al. (2005) Control of lipid metabolism by phosphorylation-dependent degradation of the SREBP family of transcription factors by SCF(Fbw7). Cell Metab 1: 379–391.

64. HughesAL, ToddBL, EspenshadePJ (2005) SREBP pathway responds to sterols and functions as an oxygen sensor in fission yeast. Cell 120: 831–842.

65. GustafssonMV, ZhengX, PereiraT, GradinK, JinS, et al. (2005) Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell 9: 617–628.

66. PerrimonN, NollE, McCallK, BrandA (1991) Generating lineage-specific markers to study Drosophila development. Dev Genet 12: 238–252.

67. JinJ, AnthopoulosN, WetschB, BinariRC, IsaacDD, et al. (2001) Regulation of Drosophila tracheal system development by protein kinase B. Dev Cell 1: 817–827.

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#