#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Cell Type-Specific Functions of Genes Revealed by Novel Adipocyte and Hepatocyte Circadian Clock Models


Various aspects of our daily rhythms in physiology and behavior such as the sleep-wake cycle are regulated by endogenous circadian clocks that are present in nearly every cell. It is generally accepted that these oscillators share a similar biochemical negative feedback mechanism, consisting of transcriptional activators and repressors. In this study, we developed cell-autonomous, metabolically relevant clock models in mouse hepatocytes and adipocytes. Each clock model has an integrated luciferase reporter that allows for kinetic luminescence recording with an inexpensive microplate reader and thus is feasible for most laboratories. These models are amenable to high throughput screening of small molecules or genomic entities for impacts on cell-autonomous clocks relevant to metabolism. We validated these new models by RNA interference via lentivirus-mediated knockdown of known clock genes. As expected, we found that many core clock components have similar functions across cell types. To our surprise, however, we also uncovered previously under-appreciated cell type-specific functions of core clock genes, particularly Per1, Per2, and Per3. Because the circadian system is integrated with, and influenced by, the local physiology that is under its control, our studies provide important implications for future studies into cell type-specific mechanisms of various circadian systems.


Vyšlo v časopise: Cell Type-Specific Functions of Genes Revealed by Novel Adipocyte and Hepatocyte Circadian Clock Models. PLoS Genet 10(4): e32767. doi:10.1371/journal.pgen.1004244
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004244

Souhrn

Various aspects of our daily rhythms in physiology and behavior such as the sleep-wake cycle are regulated by endogenous circadian clocks that are present in nearly every cell. It is generally accepted that these oscillators share a similar biochemical negative feedback mechanism, consisting of transcriptional activators and repressors. In this study, we developed cell-autonomous, metabolically relevant clock models in mouse hepatocytes and adipocytes. Each clock model has an integrated luciferase reporter that allows for kinetic luminescence recording with an inexpensive microplate reader and thus is feasible for most laboratories. These models are amenable to high throughput screening of small molecules or genomic entities for impacts on cell-autonomous clocks relevant to metabolism. We validated these new models by RNA interference via lentivirus-mediated knockdown of known clock genes. As expected, we found that many core clock components have similar functions across cell types. To our surprise, however, we also uncovered previously under-appreciated cell type-specific functions of core clock genes, particularly Per1, Per2, and Per3. Because the circadian system is integrated with, and influenced by, the local physiology that is under its control, our studies provide important implications for future studies into cell type-specific mechanisms of various circadian systems.


Zdroje

1. ReppertSM, WeaverDR (2002) Coordination of circadian timing in mammals. Nature 418: 935–941.

2. HastingsMH, ReddyAB, MaywoodES (2003) A clockwork web: circadian timing in brain and periphery, in health and disease. Nat Rev Neurosci 4: 649–661.

3. LiuAC, LewisWG, KaySA (2007) Mammalian circadian signaling networks and therapeutic targets. Nat Chem Biol 3: 630–639.

4. MohawkJA, GreenCB, TakahashiJS (2012) Central and peripheral circadian clocks in mammals. Annu Rev Neurosci 35: 445–462.

5. GreenCB, TakahashiJS, BassJ (2008) The meter of metabolism. Cell 134: 728–742.

6. NagoshiE, SainiC, BauerC, LarocheT, NaefF, et al. (2004) Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119: 693–705.

7. WelshDK, YooSH, LiuAC, TakahashiJS, KaySA (2004) Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression. Curr Biol 14: 2289–2295.

8. YooSH, YamazakiS, LowreyPL, ShimomuraK, KoCH, et al. (2004) PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc Natl Acad Sci U S A 101: 5339–5346.

9. YoungMW, KaySA (2001) Time zones: a comparative genetics of circadian clocks. Nat Rev Genet 2: 702–715.

10. BusinoL, BassermannF, MaiolicaA, LeeC, NolanPM, et al. (2007) SCFFbxl3 controls the oscillation of the circadian clock by directing the degradation of cryptochrome proteins. Science 316: 900–904.

11. SiepkaSM, YooSH, ParkJ, SongW, KumarV, et al. (2007) Circadian mutant Overtime reveals F-box protein FBXL3 regulation of cryptochrome and period gene expression. Cell 129: 1011–1023.

12. GodinhoSI, MaywoodES, ShawL, TucciV, BarnardAR, et al. (2007) The after-hours mutant reveals a role for Fbxl3 in determining mammalian circadian period. Science 316: 897–900.

13. ShiG, XingL, LiuZ, QuZ, WuX, et al. (2013) Dual roles of FBXL3 in the mammalian circadian feedback loops are important for period determination and robustness of the clock. Proc Natl Acad Sci U S A 110: 4750–4755.

14. YooSH, MohawkJA, SiepkaSM, ShanY, HuhSK, et al. (2013) Competing E3 ubiquitin ligases govern circadian periodicity by degradation of CRY in nucleus and cytoplasm. Cell 152: 1091–1105.

15. HiranoA, YumimotoK, TsunematsuR, MatsumotoM, OyamaM, et al. (2013) FBXL21 regulates oscillation of the circadian clock through ubiquitination and stabilization of cryptochromes. Cell 152: 1106–1118.

16. UedaHR, HayashiS, ChenW, SanoM, MachidaM, et al. (2005) System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat Genet 37: 187–192.

17. LiuAC, TranHG, ZhangEE, PriestAA, WelshDK, et al. (2008) Redundant function of REV-ERBalpha and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet 4: e1000023.

18. ChoH, ZhaoX, HatoriM, YuRT, BarishGD, et al. (2012) Regulation of circadian behaviour and metabolism by REV-ERB-alpha and REV-ERB-beta. Nature 485: 123–127.

19. KoikeN, YooSH, HuangHC, KumarV, LeeC, et al. (2012) Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 338: 349–354.

20. DeBruyneJP, HogeneschJB (2011) A CRY in the Night. Dev Cell 20: 144–145.

21. Ukai-TadenumaM, YamadaRG, XuH, RippergerJA, LiuAC, et al. (2011) Delay in feedback repression by cryptochrome 1 is required for circadian clock function. Cell 144: 268–281.

22. HogeneschJB, UedaHR (2011) Understanding systems-level properties: timely stories from the study of clocks. Nat Rev Genet 12: 407–416.

23. RamanathanC, KhanSK, KathaleND, XuH, LiuAC (2012) Monitoring cell-autonomous circadian clock rhythms of gene expression using luciferase bioluminescence reporters. J Vis Exp e4234 doi 4210.37914234.:4210.3791/4234.

24. KhanSK, XuH, Ukai-TadenumaM, BurtonB, WangY, et al. (2012) Identification of a novel cryptochrome differentiating domain required for feedback repression in circadian clock function. J Biol Chem 287: 25917–25926.

25. HogeneschJB, HerzogED (2011) Intracellular and intercellular processes determine robustness of the circadian clock. FEBS Lett 585: 1427–1434.

26. UkaiH, UedaHR (2010) Systems biology of mammalian circadian clocks. Annu Rev Physiol 72: 579–603.

27. BaggsJE, PriceTS, DiTacchioL, PandaS, FitzgeraldGA, et al. (2009) Network features of the mammalian circadian clock. PLoS Biol 7: e52.

28. SatoTK, YamadaRG, UkaiH, BaggsJE, MiragliaLJ, et al. (2006) Feedback repression is required for mammalian circadian clock function. Nat Genet 38: 312–319.

29. AtwoodA, DeCondeR, WangSS, MocklerTC, SabirJS, et al. (2011) Cell-autonomous circadian clock of hepatocytes drives rhythms in transcription and polyamine synthesis. Proc Natl Acad Sci U S A 108: 18560–18565.

30. PandaS, AntochMP, MillerBH, SuAI, SchookAB, et al. (2002) Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109: 307–320.

31. ZhangEE, LiuAC, HirotaT, MiragliaLJ, WelchG, et al. (2009) A genome-wide RNAi screen for modifiers of the circadian clock in human cells. Cell 139: 199–210.

32. Eckel-MahanK, Sassone-CorsiP (2009) Metabolism control by the circadian clock and vice versa. Nat Struct Mol Biol 16: 462–467.

33. AsherG, SchiblerU (2011) Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab 13: 125–137.

34. GreenH, KehindeO (1975) An established preadipose cell line and its differentiation in culture. II. Factors affecting the adipose conversion. Cell 5: 19–27.

35. KallenCB, LazarMA (1996) Antidiabetic thiazolidinediones inhibit leptin (ob) gene expression in 3T3-L1 adipocytes. Proc Natl Acad Sci U S A 93: 5793–5796.

36. AmiconeL, SpagnoliFM, SpathG, GiordanoS, TommasiniC, et al. (1997) Transgenic expression in the liver of truncated Met blocks apoptosis and permits immortalization of hepatocytes. EMBO J 16: 495–503.

37. FeigelstockDA, ThompsonP, KaplanGG (2005) Growth of hepatitis A virus in a mouse liver cell line. J Virol 79: 2950–2955.

38. OtwayDT, FrostG, JohnstonJD (2009) Circadian rhythmicity in murine pre-adipocyte and adipocyte cells. Chronobiol Int 26: 1340–1354.

39. AoyagiT, ShimbaS, TezukaM (2005) Characteristics of Circadian Gene Expression in Mice White Adipose Tissue and 3T3-L1 Adipocytes. Journal of Health Science 51: 21–32.

40. GachonF, OlelaFF, SchaadO, DescombesP, SchiblerU (2006) The circadian PAR-domain basic leucine zipper transcription factors DBP, TEF, and HLF modulate basal and inducible xenobiotic detoxification. Cell Metab 4: 25–36.

41. YamajukuD, ShibataY, KitazawaM, KatakuraT, UrataH, et al. (2011) Cellular DBP and E4BP4 proteins are critical for determining the period length of the circadian oscillator. FEBS Lett 585: 2217–2222.

42. EcheverriCJ, BeachyPA, BaumB, BoutrosM, BuchholzF, et al. (2006) Minimizing the risk of reporting false positives in large-scale RNAi screens. Nat Methods 3: 777–779.

43. LiuAC, WelshDK, KoCH, TranHG, ZhangEE, et al. (2007) Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell 129: 605–616.

44. MaierB, WendtS, VanselowJT, WallachT, ReischlS, et al. (2009) A large-scale functional RNAi screen reveals a role for CK2 in the mammalian circadian clock. Genes Dev 23: 708–718.

45. KoCH, YamadaYR, WelshDK, BuhrED, LiuAC, et al. (2010) Emergence of noise-induced oscillations in the central circadian pacemaker. PLoS Biol 8: e1000513.

46. DeBruyneJP, WeaverDR, ReppertSM (2007) Peripheral circadian oscillators require CLOCK. Curr Biol 17: R538–539.

47. DudleyCA, Erbel-SielerC, EstillSJ, ReickM, FrankenP, et al. (2003) Altered patterns of sleep and behavioral adaptability in NPAS2-deficient mice. Science 301: 379–383.

48. ShiS, HidaA, McGuinnessOP, WassermanDH, YamazakiS, et al. (2010) Circadian clock gene Bmal1 is not essential; functional replacement with its paralog, Bmal2. Curr Biol 20: 316–321.

49. PreitnerN, DamiolaF, Lopez-MolinaL, ZakanyJ, DubouleD, et al. (2002) The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 110: 251–260.

50. PendergastJS, FridayRC, YamazakiS (2009) Endogenous rhythms in Period1 mutant suprachiasmatic nuclei in vitro do not represent circadian behavior. J Neurosci 29: 14681–14686.

51. BaeK, JinX, MaywoodES, HastingsMH, ReppertSM, et al. (2001) Differential functions of mPer1, mPer2, and mPer3 in the SCN circadian clock. Neuron 30: 525–536.

52. ZhengB, AlbrechtU, KaasikK, SageM, LuW, et al. (2001) Nonredundant roles of the mPer1 and mPer2 genes in the mammalian circadian clock. Cell 105: 683–694.

53. BrownSA, Fleury-OlelaF, NagoshiE, HauserC, JugeC, et al. (2005) The period length of fibroblast circadian gene expression varies widely among human individuals. PLoS Biol 3: e338.

54. PendergastJS, FridayRC, YamazakiS (2010) Distinct functions of Period2 and Period3 in the mouse circadian system revealed by in vitro analysis. PLoS One 5: e8552.

55. ChenR, SchirmerA, LeeY, LeeH, KumarV, et al. (2009) Rhythmic PER abundance defines a critical nodal point for negative feedback within the circadian clock mechanism. Mol Cell 36: 417–430.

56. ShearmanLP, JinX, LeeC, ReppertSM, WeaverDR (2000) Targeted disruption of the mPer3 gene: subtle effects on circadian clock function. Mol Cell Biol 20: 6269–6275.

57. YagitaK, TamaniniF, van Der HorstGT, OkamuraH (2001) Molecular mechanisms of the biological clock in cultured fibroblasts. Science 292: 278–281.

58. PendergastJS, NiswenderKD, YamazakiS (2012) Tissue-specific function of Period3 in circadian rhythmicity. PLoS One 7: e30254.

59. EvansJA, PanH, LiuAC, WelshDK (2012) Cry1−/− circadian rhythmicity depends on SCN intercellular coupling. J Biol Rhythms 27: 443–452.

60. HasanS, van der VeenDR, Winsky-SommererR, DijkDJ, ArcherSN (2011) Altered sleep and behavioral activity phenotypes in PER3-deficient mice. Am J Physiol Regul Integr Comp Physiol 301: R1821–1830.

61. DallmannR, WeaverDR (2010) Altered body mass regulation in male mPeriod mutant mice on high-fat diet. Chronobiol Int 27: 1317–1328.

62. CostaMJ, SoAY, KaasikK, KruegerKC, PillsburyML, et al. (2011) Circadian rhythm gene period 3 is an inhibitor of the adipocyte cell fate. J Biol Chem 286: 9063–9070.

63. BarnardAR, NolanPM (2008) When clocks go bad: neurobehavioural consequences of disrupted circadian timing. PLoS Genet 4: e1000040.

64. DijkDJ, ArcherSN (2009) PERIOD3, circadian phenotypes, and sleep homeostasis. Sleep Med Rev 14: 151–160.

65. ShearmanLP, SriramS, WeaverDR, MaywoodES, ChavesI, et al. (2000) Interacting molecular loops in the mammalian circadian clock. Science 288: 1013–1019.

66. SchmutzI, RippergerJA, Baeriswyl-AebischerS, AlbrechtU (2010) The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev 24: 345–357.

67. KimJK, ForgerDB (2012) A mechanism for robust circadian timekeeping via stoichiometric balance. Mol Syst Biol 8: 630.

68. LeeY, ChenR, LeeHM, LeeC (2011) Stoichiometric relationship among clock proteins determines robustness of circadian rhythms. J Biol Chem 286: 7033–7042.

69. YeR, SelbyCP, OzturkN, AnnayevY, SancarA (2011) Biochemical analysis of the canonical model for the mammalian circadian clock. J Biol Chem 286: 25891–25902.

70. HartGW (2013) How sugar tunes your clock. Cell Metab 17: 155–156.

71. PaganoM, JacksonPK (2004) Wagging the dogma; tissue-specific cell cycle control in the mouse embryo. Cell 118: 535–538.

72. Aza-BlancP, CooperCL, WagnerK, BatalovS, DeverauxQL, et al. (2003) Identification of modulators of TRAIL-induced apoptosis via RNAi-based phenotypic screening. Mol Cell 12: 627–637.

73. RubinsonDA, DillonCP, KwiatkowskiAV, SieversC, YangL, et al. (2003) A lentivirus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference. Nat Genet 33: 401–406.

74. TiscorniaG, SingerO, VermaIM (2006) Production and purification of lentiviral vectors. Nat Protoc 1: 241–245.

75. HirotaT, LewisWG, LiuAC, LeeJW, SchultzPG, et al. (2008) A chemical biology approach reveals period shortening of the mammalian circadian clock by specific inhibition of GSK-3beta. Proc Natl Acad Sci U S A 105: 20746–20751.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 4
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#