#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

TATN-1 Mutations Reveal a Novel Role for Tyrosine as a Metabolic Signal That Influences Developmental Decisions and Longevity in


Recent work has identified changes in the metabolism of the aromatic amino acid tyrosine as a risk factor for diabetes and a contributor to the development of liver cancer. While these findings could suggest a role for tyrosine as a direct regulator of the behavior of cells and tissues, evidence for this model is currently lacking. Through the use of RNAi and genetic mutants, we identify tatn-1, which is the worm ortholog of tyrosine aminotransferase and catalyzes the first step of the conserved tyrosine degradation pathway, as a novel regulator of the dauer decision and modulator of the daf-2 insulin/IGF-1-like (IGFR) signaling pathway in Caenorhabditis elegans. Mutations affecting tatn-1 elevate tyrosine levels in the animal, and enhance the effects of mutations in genes that lie within the daf-2/insulin signaling pathway or are otherwise upstream of daf-16/FOXO on both dauer formation and worm longevity. These effects are mediated by elevated tyrosine levels as supplemental dietary tyrosine mimics the phenotypes produced by a tatn-1 mutation, and the effects still occur when the enzymes needed to convert tyrosine into catecholamine neurotransmitters are missing. The effects on dauer formation and lifespan require the aak-2/AMPK gene, and tatn-1 mutations increase phospho-AAK-2 levels. In contrast, the daf-16/FOXO transcription factor is only partially required for the effects on dauer formation and not required for increased longevity. We also find that the controlled metabolism of tyrosine by tatn-1 may function normally in dauer formation because the expression of the TATN-1 protein is regulated both by daf-2/IGFR signaling and also by the same dietary and environmental cues which influence dauer formation. Our findings point to a novel role for tyrosine as a developmental regulator and modulator of longevity, and support a model where elevated tyrosine levels play a causal role in the development of diabetes and cancer in people.


Vyšlo v časopise: TATN-1 Mutations Reveal a Novel Role for Tyrosine as a Metabolic Signal That Influences Developmental Decisions and Longevity in. PLoS Genet 9(12): e32767. doi:10.1371/journal.pgen.1004020
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004020

Souhrn

Recent work has identified changes in the metabolism of the aromatic amino acid tyrosine as a risk factor for diabetes and a contributor to the development of liver cancer. While these findings could suggest a role for tyrosine as a direct regulator of the behavior of cells and tissues, evidence for this model is currently lacking. Through the use of RNAi and genetic mutants, we identify tatn-1, which is the worm ortholog of tyrosine aminotransferase and catalyzes the first step of the conserved tyrosine degradation pathway, as a novel regulator of the dauer decision and modulator of the daf-2 insulin/IGF-1-like (IGFR) signaling pathway in Caenorhabditis elegans. Mutations affecting tatn-1 elevate tyrosine levels in the animal, and enhance the effects of mutations in genes that lie within the daf-2/insulin signaling pathway or are otherwise upstream of daf-16/FOXO on both dauer formation and worm longevity. These effects are mediated by elevated tyrosine levels as supplemental dietary tyrosine mimics the phenotypes produced by a tatn-1 mutation, and the effects still occur when the enzymes needed to convert tyrosine into catecholamine neurotransmitters are missing. The effects on dauer formation and lifespan require the aak-2/AMPK gene, and tatn-1 mutations increase phospho-AAK-2 levels. In contrast, the daf-16/FOXO transcription factor is only partially required for the effects on dauer formation and not required for increased longevity. We also find that the controlled metabolism of tyrosine by tatn-1 may function normally in dauer formation because the expression of the TATN-1 protein is regulated both by daf-2/IGFR signaling and also by the same dietary and environmental cues which influence dauer formation. Our findings point to a novel role for tyrosine as a developmental regulator and modulator of longevity, and support a model where elevated tyrosine levels play a causal role in the development of diabetes and cancer in people.


Zdroje

1. NewgardCB, AnJ, BainJR, MuehlbauerMJ, StevensRD, et al. (2009) A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab 9: 311–326.

2. WurtzP, TiainenM, MakinenVP, KangasAJ, SoininenP, et al. (2012) Circulating Metabolite Predictors of Glycemia in Middle-Aged Men and Women. Diabetes Care

3. WangTJ, LarsonMG, VasanRS, ChengS, RheeEP, et al. (2011) Metabolite profiles and the risk of developing diabetes. Nat Med 17: 448–453.

4. StancakovaA, CivelekM, SaleemNK, SoininenP, KangasAJ, et al. (2012) Hyperglycemia and a Common Variant of GCKR Are Associated With the Levels of Eight Amino Acids in 9,369 Finnish Men. Diabetes

5. ChengS, RheeEP, LarsonMG, LewisGD, McCabeEL, et al. (2012) Metabolite profiling identifies pathways associated with metabolic risk in humans. Circulation 125: 2222–2231.

6. WurtzP, MakinenVP, SoininenP, KangasAJ, TukiainenT, et al. (2012) Metabolic signatures of insulin resistance in 7,098 young adults. Diabetes 61: 1372–1380.

7. FuL, DongSS, XieYW, TaiLS, ChenL, et al. (2010) Down-regulation of tyrosine aminotransferase at a frequently deleted region 16q22 contributes to the pathogenesis of hepatocellular carcinoma. Hepatology 51: 1624–1634.

8. CassadaRC, RussellRL (1975) The dauerlarva, a post-embryonic developmental variant of the nematode Caenorhabditis elegans. Dev Biol 46: 326–342.

9. Riddle DL, Albert PS, Riddle DL, Blumenthal T, Meyer BJ, et al.. (1997) Genetic and environmental regulation of dauer larva formation. C elegans II. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press. pp. 739–768.

10. HuPJ (2007) Dauer. WormBook 1–19.

11. FielenbachN, AntebiA (2008) C. elegans dauer formation and the molecular basis of plasticity. Genes Dev 22: 2149–2165.

12. RiddleDL, SwansonMM, AlbertPS (1981) Interacting genes in nematode dauer larva formation. Nature 290: 668–671.

13. KenyonC, ChangJ, GenschE, RudnerA, TabtiangR (1993) A C. elegans mutant that lives twice as long as wild type. Nature 366: 461–464.

14. KimuraKD, TissenbaumHA, LiuY, RuvkunG (1997) daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science 277: 942–946.

15. PierceSB, CostaM, WisotzkeyR, DevadharS, HomburgerSA, et al. (2001) Regulation of DAF-2 receptor signaling by human insulin and ins-1, a member of the unusually large and diverse C. elegans insulin gene family. Genes and Development 15: 672–686.

16. LiW, KennedySG, RuvkunG (2003) daf-28 encodes a C. elegans insulin superfamily member that is regulated by environmental cues and acts in the DAF-2 signaling pathway. Genes Dev 17: 844–858.

17. MichaelsonD, KortaDZ, CapuaY, HubbardEJ (2010) Insulin signaling promotes germline proliferation in C. elegans. Development 137: 671–680.

18. CornilsA, GloeckM, ChenZ, ZhangY, AlcedoJ (2011) Specific insulin-like peptides encode sensory information to regulate distinct developmental processes. Development 138: 1183–1193.

19. MaloneEA, InoueT, ThomasJH (1996) Genetic analysis of the roles of daf-28 and age-1 in regulating Caenorhabditis elegans dauer formation. Genetics 143: 1193–1205.

20. ParadisS, RuvkunG (1998) Caenorhabditis elegans Akt/PKB transduces insulin receptor-like signals from AGE-1 PI3 kinase to the DAF-16 transcription factor. Genes and Development 12: 2488–2498.

21. ParadisS, AilionM, TokerA, ThomasJH, RuvkunG (1999) A PDK1 homolog is necessary and sufficient to transduce AGE-1 PI3 kinase signals that regulate diapause in Caenorhabditis elegans. Genes and Development 13: 1438–1452.

22. MorrisJZ, TissenbaumHA, RuvkunG (1996) A phosphatidylinositol-3-OH kinase family member regulating longevity and diapause in Caenorhabditis elegans. Nature 382: 536–539.

23. LinK, HsinH, LibinaN, KenyonC (2001) Regulation of the Caenorhabditis elegans longevity protein DAF-16 by insulin/IGF-1 and germline signaling. NatGenet 28: 139–145.

24. LeeRY, HenchJ, RuvkunG (2001) Regulation of C. elegans DAF-16 and its human ortholog FKHRL1 by the daf-2 insulin-like signaling pathway. CurrBiol 11: 1950–1957.

25. HendersonST, JohnsonTE (2001) daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. CurrBiol 11: 1975–1980.

26. ZhangY, XuJ, PuscauC, KimY, WangX, et al. (2008) Caenorhabditis elegans EAK-3 inhibits dauer arrest via nonautonomous regulation of nuclear DAF-16/FoxO activity. Dev Biol 315: 290–302.

27. AlamH, WilliamsTW, DumasKJ, GuoC, YoshinaS, et al. (2010) EAK-7 controls development and life span by regulating nuclear DAF-16/FoxO activity. Cell Metab 12: 30–41.

28. HuPJ, XuJ, RuvkunG (2006) Two membrane-associated tyrosine phosphatase homologs potentiate C. elegans AKT-1/PKB signaling. PLoS Genet 2: e99.

29. DumasKJ, GuoC, WangX, BurkhartKB, AdamsEJ, et al. (2010) Functional divergence of dafachronic acid pathways in the control of C. elegans development and lifespan. Dev Biol 340: 605–612.

30. SpencerCJ, HeatonJH, GelehrterTD, RichardsonKI, GarwinJL (1978) Insulin selectively slows the degradation rate of tyrosine aminotransferase. J Biol Chem 253: 7677–7682.

31. ReshefL, GreengardO (1969) The effect of amino acid mixtures, insulin, epinephrine and glucagon in vivo on the levels of rat liver tyrosine aminotransferase. Enzymol Biol Clin (Basel) 10: 113–121.

32. NitschD, BoshartM, SchutzG (1993) Activation of the tyrosine aminotransferase gene is dependent on synergy between liver-specific and hormone-responsive elements. Proc Natl Acad Sci U S A 90: 5479–5483.

33. MoorePS, KoontzJW (1989) Insulin-mediated regulation of tyrosine aminotransferase in rat hepatoma cells: inhibition of transcription and inhibition of enzyme degradation. Arch Biochem Biophys 275: 486–495.

34. MessinaJL, ChatterjeeAK, StrapkoHT, WeinstockRS (1992) Short- and long-term effects of insulin on tyrosine aminotransferase gene expression. Arch Biochem Biophys 298: 56–62.

35. LeeKL, IshamKR, JohnsonA, KenneyFT (1986) Insulin enhances transcription of the tyrosine aminotransferase gene in rat liver. Arch Biochem Biophys 248: 597–603.

36. GelehrterTD, TomkinsGM (1970) Posttranscriptional control of tyrosine aminotransferase synthesis by insulin. Proc Natl Acad Sci U S A 66: 390–397.

37. GelehrterTD, EmanuelJR, SpencerCJ (1972) Induction of tyrosine aminotransferase by dexamethasone, insulin, and serum. Characterization of the induced enzyme. J Biol Chem 247: 6197–6203.

38. CrettazM, Muller-WielandD, KahnCR (1988) Transcriptional and posttranscriptional regulation of tyrosine aminotransferase by insulin in rat hepatoma cells. Biochemistry 27: 495–500.

39. LeeSS, KennedyS, TolonenAC, RuvkunG (2003) DAF-16 target genes that control C. elegans life-span and metabolism. Science 300: 644–647.

40. FuchsS, BundyJG, DaviesSK, VineyJM, SwireJS, et al. (2010) A metabolic signature of long life in Caenorhabditis elegans. BMC Biol 8: 14.

41. TewariM, HuPJ, AhnJS, yivi-GuedehoussouN, VidalainPO, et al. (2004) Systematic interactome mapping and genetic perturbation analysis of a C. elegans TGF-beta signaling network. MolCell 13: 469–482.

42. DuchaineTF, WohlschlegelJA, KennedyS, BeiY, ConteDJr, et al. (2006) Functional proteomics reveals the biochemical niche of C. elegans DCR-1 in multiple small-RNA-mediated pathways. Cell 124: 343–354.

43. FisherAL, PageKE, LithgowGJ, NashL (2008) The Caenorhabditis elegans K10C2.4 Gene Encodes a Member of the Fumarylacetoacetate Hydrolase Family: A CAENORHABDITIS ELEGANS MODEL OF TYPE I TYROSINEMIA. J BiolChem 283: 9127–9135.

44. FergusonAA, SpringerMG, FisherAL (2010) skn-1-Dependent and -independent regulation of aip-1 expression following metabolic stress in Caenorhabditis elegans. Mol Cell Biol 30: 2651–2667.

45. HertweckM, GobelC, BaumeisterR (2004) C. elegans SGK-1 is the critical component in the Akt/PKB kinase complex to control stress response and life span. Dev Cell 6: 577–588.

46. JensenVL, AlbertPS, RiddleDL (2007) Caenorhabditis elegans SDF-9 enhances insulin/insulin-like signaling through interaction with DAF-2. Genetics 177: 661–666.

47. NarasimhanSD, YenK, BansalA, KwonES, PadmanabhanS, et al. (2011) PDP-1 links the TGF-beta and IIS pathways to regulate longevity, development, and metabolism. PLoS Genet 7: e1001377.

48. PattersonGI, KoweekA, WongA, LiuY, RuvkunG (1997) The DAF-3 Smad protein antagonizes TGF-beta-related receptor signaling in the Caenorhabditis elegans dauer pathway. Genes Dev 11: 2679–2690.

49. da GracaLS, ZimmermanKK, MitchellMC, Kozhan-GorodetskaM, SekiewiczK, et al. (2004) DAF-5 is a Ski oncoprotein homolog that functions in a neuronal TGF beta pathway to regulate C. elegans dauer development. Development 131: 435–446.

50. ApfeldJ, O'ConnorG, McDonaghT, DiStefanoPS, CurtisR (2004) The AMP-activated protein kinase AAK-2 links energy levels and insulin-like signals to lifespan in C. elegans. Genes Dev 18: 3004–3009.

51. GreerEL, DowlatshahiD, BankoMR, VillenJ, HoangK, et al. (2007) An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol 17: 1646–1656.

52. NarbonneP, RoyR (2009) Caenorhabditis elegans dauers need LKB1/AMPK to ration lipid reserves and ensure long-term survival. Nature 457: 210–214.

53. MairW, MorantteI, RodriguesAP, ManningG, MontminyM, et al. (2011) Lifespan extension induced by AMPK and calcineurin is mediated by CRTC-1 and CREB. Nature 470: 404–408.

54. LagidoC, PettittJ, FlettA, GloverLA (2008) Bridging the phenotypic gap: real-time assessment of mitochondrial function and metabolism of the nematode Caenorhabditis elegans. BMC Physiol 8: 7.

55. KwonES, NarasimhanSD, YenK, TissenbaumHA (2010) A new DAF-16 isoform regulates longevity. Nature 466: 498–502.

56. Robida-StubbsS, Glover-CutterK, LammingDW, MizunumaM, NarasimhanSD, et al. (2012) TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metab 15: 713–724.

57. HondaY, HondaS (1999) The daf-2 gene network for longevity regulates oxidative stress resistance and Mn-superoxide dismutase gene expression in Caenorhabditis elegans. FASEB J 13: 1385–1393.

58. LibinaN, BermanJR, KenyonC (2003) Tissue-specific activities of C. elegans DAF-16 in the regulation of lifespan. Cell 115: 489–502.

59. BermanJR, KenyonC (2006) Germ-cell loss extends C. elegans life span through regulation of DAF-16 by kri-1 and lipophilic-hormone signaling. Cell 124: 1055–1068.

60. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

61. MiH, MuruganujanA, ThomasPD (2013) PANTHER in 2013: modeling the evolution of gene function, and other gene attributes, in the context of phylogenetic trees. Nucleic Acids Res 41: D377–386.

62. MurphyCT, McCarrollSA, BargmannCI, FraserA, KamathRS, et al. (2003) Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature 424: 277–283.

63. XiongQ, AnconaN, HauserER, MukherjeeS, FureyTS (2012) Integrating genetic and gene expression evidence into genome-wide association analysis of gene sets. Genome Res 22: 386–397.

64. BrooksKK, LiangB, WattsJL (2009) The influence of bacterial diet on fat storage in C. elegans. PLoS ONE 4: e7545.

65. ZigmondMJ, ShoemakerWJ, LarinF, WurtmanRJ (1969) Hepatic tyrosine transaminase rhythm: interaction of environmental lighting, food consumption and dietary protein content. J Nutr 98: 71–75.

66. WurtmanRJ (1974) Daily rhythms in tyrosine transaminase and other hepatic enzymes that metabolize amino acids: mechanisms and possible consequences. Life Sci 15: 827–847.

67. RossDS, FernstromJD, WurtmanRJ (1973) The role of dietary protein in generating daily rhythms in rat liver tryptophan pyrrolase and tyrosine transaminase. Metabolism 22: 1175–1184.

68. IsaacsJS, JungYJ, MoleDR, LeeS, Torres-CabalaC, et al. (2005) HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell 8: 143–153.

69. CitronBA, DavisMD, MilstienS, GutierrezJ, MendelDB, et al. (1992) Identity of 4a-carbinolamine dehydratase, a component of the phenylalanine hydroxylation system, and DCoH, a transregulator of homeodomain proteins. ProcNatlAcadSciUSA 89: 11891–11894.

70. CalvoAC, PeyAL, YingM, LoerCM, MartinezA (2008) Anabolic function of phenylalanine hydroxylase in Caenorhabditis elegans. FASEB J 22: 3046–3058.

71. TremblayF, LavigneC, JacquesH, MaretteA (2007) Role of dietary proteins and amino acids in the pathogenesis of insulin resistance. Annu Rev Nutr 27: 293–310.

72. TremblayF, MaretteA (2001) Amino acid and insulin signaling via the mTOR/p70 S6 kinase pathway. A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J Biol Chem 276: 38052–38060.

73. WurtmanRJ, LarinF, MostafapourS, FernstromJD (1974) Brain catechol synthesis: control by train tyrosine concentration. Science 185: 183–184.

74. LintsR, EmmonsSW (1999) Patterning of dopaminergic neurotransmitter identity among Caenorhabditis elegans ray sensory neurons by a TGFbeta family signaling pathway and a Hox gene. Development 126: 5819–5831.

75. AlkemaMJ, Hunter-EnsorM, RingstadN, HorvitzHR (2005) Tyramine Functions independently of octopamine in the Caenorhabditis elegans nervous system. Neuron 46: 247–260.

76. GreerEL, OskouiPR, BankoMR, ManiarJM, GygiMP, et al. (2007) The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J Biol Chem 282: 30107–30119.

77. KooSH, FlechnerL, QiL, ZhangX, ScreatonRA, et al. (2005) The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 437: 1109–1111.

78. DentinR, LiuY, KooSH, HedrickS, VargasT, et al. (2007) Insulin modulates gluconeogenesis by inhibition of the coactivator TORC2. Nature 449: 366–369.

79. CypserJR, JohnsonTE (2002) Multiple stressors in Caenorhabditis elegans induce stress hormesis and extended longevity. J Gerontol A Biol Sci Med Sci 57: B109–114.

80. WalkerGA, ThompsonFJ, BrawleyA, ScanlonT, DevaneyE (2003) Heat shock factor functions at the convergence of the stress response and developmental pathways in Caenorhabditis elegans. FASEB J 17: 1960–1962.

81. MorleyJF, MorimotoRI (2004) Regulation of longevity in Caenorhabditis elegans by heat shock factor and molecular chaperones. Mol Biol Cell 15: 657–664.

82. HsuAL, MurphyCT, KenyonC (2003) Regulation of aging and age-related disease by DAF-16 and heat-shock factor. Science 300: 1142–1145.

83. ConigraveAD, QuinnSJ, BrownEM (2000) L-amino acid sensing by the extracellular Ca2+-sensing receptor. Proc Natl Acad Sci U S A 97: 4814–4819.

84. BittingerMA, NguyenLP, BradfieldCA (2003) Aspartate aminotransferase generates proagonists of the aryl hydrocarbon receptor. Mol Pharmacol 64: 550–556.

85. SchumacherU, LukacsZ, KaltschmidtC, FreudlspergerC, SchulzD, et al. (2008) High concentrations of phenylalanine stimulate peroxisome proliferator-activated receptor gamma: implications for the pathophysiology of phenylketonuria. Neurobiol Dis 32: 385–390.

86. LuM, WanM, LeavensKF, ChuQ, MonksBR, et al. (2012) Insulin regulates liver metabolism in vivo in the absence of hepatic Akt and Foxo1. Nat Med 18: 388–395.

87. WurtmanRJ, AxelrodJ (1967) Daily rhythmic changes in tyrosine transaminase activity of the rat liver. Proc Natl Acad Sci U S A 57: 1594–1598.

88. LeeSJ, KenyonC (2009) Regulation of the longevity response to temperature by thermosensory neurons in Caenorhabditis elegans. Curr Biol 19: 715–722.

89. LangenbergC, SavageDB (2011) An amino acid profile to predict diabetes? Nat Med 17: 418–420.

90. Sulston JE, Horvitz HR (1988) Methods. In: Wood WB, editor. The nematode, Caenorhabditis elegans. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press. pp. 587–606.

91. PatelDS, FangLL, SvyDK, RuvkunG, LiW (2008) Genetic identification of HSD-1, a conserved steroidogenic enzyme that directs larval development in Caenorhabditis elegans. Development 135: 2239–2249.

92. LandisJR, KochGG (1977) The measurement of observer agreement for categorical data. Biometrics 33: 159–174.

93. PowolnyAA, SinghSV, MelovS, HubbardA, FisherAL (2011) The garlic constituent diallyl trisulfide increases the lifespan of C. elegans via skn-1 activation. Exp Gerontol

94. GeierFM, WantEJ, LeroiAM, BundyJG (2011) Cross-platform comparison of Caenorhabditis elegans tissue extraction strategies for comprehensive metabolome coverage. Anal Chem 83: 3730–3736.

95. HeldPK, WhiteL, PasqualiM (2011) Quantitative urine amino acid analysis using liquid chromatography tandem mass spectrometry and aTRAQ reagents. J Chromatogr B Analyt Technol Biomed Life Sci 879: 2695–2703.

96. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.

97. HoogewijsD, HouthoofdK, MatthijssensF, VandesompeleJ, VanfleterenJR (2008) Selection and validation of a set of reliable reference genes for quantitative sod gene expression analysis in C. elegans. BMC Mol Biol 9: 9.

98. HochbaumD, ZhangY, StuckenholzC, LabhartP, AlexiadisV, et al. (2011) DAF-12 regulates a connected network of genes to ensure robust developmental decisions. PLoS Genet 7: e1002179.

99. LiJ, EbataA, DongY, RizkiG, IwataT, et al. (2008) Caenorhabditis elegans HCF-1 functions in longevity maintenance as a DAF-16 regulator. PLoS Biol 6: e233.

100. MarshallOJ (2004) PerlPrimer: cross-platform, graphical primer design for standard, bisulphite and real-time PCR. Bioinformatics 20: 2471–2472.

101. LiB, DeweyCN (2011) RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12: 323.

102. BullardJH, PurdomE, HansenKD, DudoitS (2010) Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics 11: 94.

103. LameschP, MilsteinS, HaoT, RosenbergJ, LiN, et al. (2004) C. elegans ORFeome version 3.1: increasing the coverage of ORFeome resources with improved gene predictions. Genome Res 14: 2064–2069.

104. DupuyD, LiQR, DeplanckeB, BoxemM, HaoT, et al. (2004) A first version of the Caenorhabditis elegans Promoterome. Genome Res 14: 2169–2175.

105. HochbaumD, FergusonAA, FisherAL (2010) Generation of transgenic C. elegans by biolistic transformation. J Vis Exp

106. FergusonAA, CaiL, KashyapL, FisherAL (2013) Improved Vectors for Selection of Transgenic Caenorhabditis elegans. Methods Mol Biol 940: 87–102.

107. ZaninE, DumontJ, GassmannR, CheesemanI, MaddoxP, et al. (2011) Affinity purification of protein complexes in C. elegans. Methods Cell Biol 106: 289–322.

108. Abramoff MDMPJ (2004) RamSJ (2004) Image Processing with ImageJ. Biophotonics International 11: 36–42.

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#