#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Uncovering Enhancer Functions Using the α-Globin Locus


Over the last three decades, studies of the α- and β-globin genes clusters have led to elucidation of the general principles of mammalian gene regulation, such as RNA stability, termination of transcription, and, more importantly, the identification of remote regulatory elements. More recently, detailed studies of α-globin regulation, using both mouse and human loci, allowed the dissection of the sequential order in which transcription factors are recruited to the locus during lineage specification. These studies demonstrated the importance of the remote regulatory elements in the recruitment of RNA polymerase II (PolII) together with their role in the generation of intrachromosomal loops within the locus and the removal of polycomb complexes during differentiation. The multiple roles attributed to remote regulatory elements that have emerged from these studies will be discussed.


Vyšlo v časopise: Uncovering Enhancer Functions Using the α-Globin Locus. PLoS Genet 10(10): e32767. doi:10.1371/journal.pgen.1004668
Kategorie: Review
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004668

Souhrn

Over the last three decades, studies of the α- and β-globin genes clusters have led to elucidation of the general principles of mammalian gene regulation, such as RNA stability, termination of transcription, and, more importantly, the identification of remote regulatory elements. More recently, detailed studies of α-globin regulation, using both mouse and human loci, allowed the dissection of the sequential order in which transcription factors are recruited to the locus during lineage specification. These studies demonstrated the importance of the remote regulatory elements in the recruitment of RNA polymerase II (PolII) together with their role in the generation of intrachromosomal loops within the locus and the removal of polycomb complexes during differentiation. The multiple roles attributed to remote regulatory elements that have emerged from these studies will be discussed.


Zdroje

1. OrkinSH (1995) Hematopoiesis: how does it happen? Curr Opin Cell Biol 7: 870–877.

2. PartingtonKM, JenkinsonEJ, AndersonG (1999) A novel method of cell separation based on dual parameter immunomagnetic cell selection. J Immunol Methods 223: 195–205.

3. EckertR, MixE, von BroenB (1976) Studies on column fractionation of immune cells. VI. Separation of thymus-derived lymphocytes by means of protein beads coated with antigen-antibody complexes. Acta Biol Med Ger 35: 663–671.

4. Vernimmen D (2014) The Molecular Basis of Normal Erythroid/Megakaryocyte Development and Mechanisms of Epigenetic/Transcriptional Deregulation Leading to Erythroleukemia and Thalassaemia. In: Bonifer C, Cockerill PN, editors. Transcriptional and Epigenetic Mechanisms Regulating Normal and Aberrant Blood Cell Development Epigenetics and Human Health. Heidelberg: Springer. pp. 247–266.

5. AnguitaE, HughesJ, HeyworthC, BlobelGA, WoodWG, et al. (2004) Globin gene activation during haemopoiesis is driven by protein complexes nucleated by GATA-1 and GATA-2. EMBO J 23: 2841–2852.

6. DrissenR, PalstraRJ, GillemansN, SplinterE, GrosveldF, et al. (2004) The active spatial organization of the beta-globin locus requires the transcription factor EKLF. Genes Dev 18: 2485–2490.

7. VernimmenD, De GobbiM, Sloane-StanleyJA, WoodWG, HiggsDR (2007) Long-range chromosomal interactions regulate the timing of the transition between poised and active gene expression. EMBO J 26: 2041–2051.

8. FrommM, BergP (1983) Simian virus 40 early- and late-region promoter functions are enhanced by the 72-base-pair repeat inserted at distant locations and inverted orientations. Mol Cell Biol 3: 991–999.

9. MoreauP, HenR, WasylykB, EverettR, GaubMP, et al. (1981) The SV40 72 base repair repeat has a striking effect on gene expression both in SV40 and other chimeric recombinants. Nucleic Acids Res 9: 6047–6068.

10. BanerjiJ, RusconiS, SchaffnerW (1981) Expression of a beta-globin gene is enhanced by remote SV40 DNA sequences. Cell 27: 299–308.

11. GrosschedlR, BirnstielML (1980) Spacer DNA sequences upstream of the T-A-T-A-A-A-T-A sequence are essential for promotion of H2A histone gene transcription in vivo. Proc Natl Acad Sci U S A 77: 7102–7106.

12. VernimmenD, BegonD, SalvadorC, GofflotS, GrooteclaesM, et al. (2003) Identification of HTF (HER2 transcription factor) as an AP-2 (activator protein-2) transcription factor and contribution of the HTF binding site to ERBB2 gene overexpression. Biochem J 370: 323–329.

13. HniszD, AbrahamBJ, LeeTI, LauA, Saint-AndreV, et al. (2013) Super-enhancers in the control of cell identity and disease. Cell 155: 934–947.

14. VoN, GoodmanRH (2001) CREB-binding protein and p300 in transcriptional regulation. J Biol Chem 276: 13505–13508.

15. CareyM (1998) The enhanceosome and transcriptional synergy. Cell 92: 5–8.

16. NoonanJP, McCallionAS (2010) Genomics of long-range regulatory elements. Annu Rev Genomics Hum Genet 11: 1–23.

17. ShiJ, WhyteWA, Zepeda-MendozaCJ, MilazzoJP, ShenC, et al. (2013) Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev 27: 2648–2662.

18. AmanoT, SagaiT, TanabeH, MizushinaY, NakazawaH, et al. (2009) Chromosomal dynamics at the Shh locus: limb bud-specific differential regulation of competence and active transcription. Dev Cell 16: 47–57.

19. HughesJR, ChengJF, VentressN, PrabhakarS, ClarkK, et al. (2005) Annotation of cis-regulatory elements by identification, subclassification, and functional assessment of multispecies conserved sequences. Proc Natl Acad Sci U S A 102: 9830–9835.

20. GohSH, LeeYT, BhanuNV, CamMC, DesperR, et al. (2005) A newly discovered human alpha-globin gene. Blood 106: 1466–1472.

21. HiggsDR (2013) The molecular basis of alpha-thalassemia. Cold Spring Harb Perspect Med 3: a011718.

22. KioussisD, VaninE, deLangeT, FlavellRA, GrosveldFG (1983) Beta-globin gene inactivation by DNA translocation in gamma beta-thalassaemia. Nature 306: 662–666.

23. GrosveldF, van AssendelftGB, GreavesDR, KolliasG (1987) Position-independent, high-level expression of the human beta-globin gene in transgenic mice. Cell 51: 975–985.

24. HattonCS, WilkieAO, DrysdaleHC, WoodWG, VickersMA, et al. (1990) Alpha-thalassemia caused by a large (62 kb) deletion upstream of the human alpha globin gene cluster. Blood 76: 221–227.

25. GalanelloR, CaoA (2011) Gene test review. Alpha-thalassemia. Genet Med 13: 83–88.

26. HiggsDR, VernimmenD, WoodB (2008) Long-range regulation of alpha-globin gene expression. Adv Genet 61: 143–173.

27. BernetA, SabatierS, PickettsDJ, OuazanaR, MorleF, et al. (1995) Targeted inactivation of the major positive regulatory element (HS-40) of the human alpha-globin gene locus. Blood 86: 1202–1211.

28. HiggsDR, WoodWG, JarmanAP, SharpeJ, LidaJ, et al. (1990) A major positive regulatory region located far upstream of the human alpha-globin gene locus. Genes Dev 4: 1588–1601.

29. GanisJJ, HsiaN, TrompoukiE, de JongJL, DiBiaseA, et al. (2012) Zebrafish globin switching occurs in two developmental stages and is controlled by the LCR. Dev Biol 366: 185–194.

30. GillemansN, McMorrowT, TewariR, WaiAW, BurgtorfC, et al. (2003) Functional and comparative analysis of globin loci in pufferfish and humans. Blood 101: 2842–2849.

31. HardisonR (1998) Hemoglobins from bacteria to man: evolution of different patterns of gene expression. J Exp Biol 201: 1099–1117.

32. JarmanAP, WoodWG, SharpeJA, GourdonG, AyyubH, et al. (1991) Characterization of the major regulatory element upstream of the human alpha-globin gene cluster. Mol Cell Biol 11: 4679–4689.

33. SharpeJA, WellsDJ, WhitelawE, VyasP, HiggsDR, et al. (1993) Analysis of the human alpha-globin gene cluster in transgenic mice. Proc Natl Acad Sci U S A 90: 11262–11266.

34. SharpeJA, SummerhillRJ, VyasP, GourdonG, HiggsDR, et al. (1993) Role of upstream DNase I hypersensitive sites in the regulation of human alpha globin gene expression. Blood 82: 1666–1671.

35. AzuaraV, PerryP, SauerS, SpivakovM, JorgensenHF, et al. (2006) Chromatin signatures of pluripotent cell lines. Nat Cell Biol 8: 532–538.

36. BernsteinBE, MikkelsenTS, XieX, KamalM, HuebertDJ, et al. (2006) A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125: 315–326.

37. SawarkarR, ParoR (2010) Interpretation of developmental signaling at chromatin: the Polycomb perspective. Dev Cell 19: 651–661.

38. SurfaceLE, ThorntonSR, BoyerLA (2010) Polycomb group proteins set the stage for early lineage commitment. Cell Stem Cell 7: 288–298.

39. MikkelsenTS, KuM, JaffeDB, IssacB, LiebermanE, et al. (2007) Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448: 553–560.

40. LongHK, BlackledgeNP, KloseRJ (2013) ZF-CxxC domain-containing proteins, CpG islands and the chromatin connection. Biochem Soc Trans 41: 727–740.

41. ClouaireT, WebbS, SkeneP, IllingworthR, KerrA, et al. (2012) Cfp1 integrates both CpG content and gene activity for accurate H3K4me3 deposition in embryonic stem cells. Genes Dev 26: 1714–1728.

42. MendenhallEM, KocheRP, TruongT, ZhouVW, IssacB, et al. (2010) GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet 6: e1001244.

43. LynchMD, SmithAJ, De GobbiM, FlenleyM, HughesJR, et al. (2011) An interspecies analysis reveals a key role for unmethylated CpG dinucleotides in vertebrate Polycomb complex recruitment. EMBO J 31: 317–329.

44. ThomsonJP, SkenePJ, SelfridgeJ, ClouaireT, GuyJ, et al. (2010) CpG islands influence chromatin structure via the CpG-binding protein Cfp1. Nature 464: 1082–1086.

45. MargueronR, JustinN, OhnoK, SharpeML, SonJ, et al. (2009) Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461: 762–767.

46. WamstadJA, AlexanderJM, TrutyRM, ShrikumarA, LiF, et al. (2012) Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151: 206–220.

47. GarrickD, De GobbiM, SamaraV, RuglessM, HollandM, et al. (2008) The role of the polycomb complex in silencing alpha-globin gene expression in nonerythroid cells. Blood 112: 3889–3899.

48. De GobbiM, GarrickD, LynchM, VernimmenD, HughesJR, et al. (2011) Generation of bivalent chromatin domains during cell fate decisions. Epigenetics Chromatin 4: 9.

49. VoigtP, TeeWW, ReinbergD (2013) A double take on bivalent promoters. Genes Dev 27: 1318–1338.

50. LevingsPP, ZhouZ, VieiraKF, Crusselle-DavisVJ, BungertJ (2006) Recruitment of transcription complexes to the beta-globin locus control region and transcription of hypersensitive site 3 prior to erythroid differentiation of murine embryonic stem cells. FEBS J 273: 746–755.

51. SzutoriszH, CanzonettaC, GeorgiouA, ChowCM, ToraL, et al. (2005) Formation of an active tissue-specific chromatin domain initiated by epigenetic marking at the embryonic stem cell stage. Mol Cell Biol 25: 1804–1820.

52. SpooncerE, HeyworthCM, DunnA, DexterTM (1986) Self-renewal and differentiation of interleukin-3-dependent multipotent stem cells are modulated by stromal cells and serum factors. Differentiation 31: 111–118.

53. GrassJA, BoyerME, PalS, WuJ, WeissMJ, et al. (2003) GATA-1-dependent transcriptional repression of GATA-2 via disruption of positive autoregulation and domain-wide chromatin remodeling. Proc Natl Acad Sci U S A 100: 8811–8816.

54. MarksPA, ShefferyM, RifkindRA (1987) Induction of transformed cells to terminal differentiation and the modulation of gene expression. Cancer Res 47: 659–666.

55. RomierC, CocchiarellaF, MantovaniR, MorasD (2003) The NF-YB/NF-YC structure gives insight into DNA binding and transcription regulation by CCAAT factor NF-Y. J Biol Chem 278: 1336–1345.

56. GattaR, MantovaniR (2008) NF-Y substitutes H2A-H2B on active cell-cycle promoters: recruitment of CoREST-KDM1 and fine-tuning of H3 methylations. Nucleic Acids Res 36: 6592–6607.

57. FunnellAP, VernimmenD, LimWF, MakKS, WienertB, et al. (2014) Differential regulation of the alpha-globin locus by Kruppel-like factor 3 in erythroid and non-erythroid cells. BMC Mol Biol 15: 8.

58. LowerKM, HughesJR, De GobbiM, HendersonS, ViprakasitV, et al. (2009) Adventitious changes in long-range gene expression caused by polymorphic structural variation and promoter competition. Proc Natl Acad Sci U S A 106: 21771–21776.

59. KowalczykMS, HughesJR, GarrickD, LynchMD, SharpeJA, et al. (2012) Intragenic enhancers act as alternative promoters. Mol Cell 45: 447–458.

60. KochF, FenouilR, GutM, CauchyP, AlbertTK, et al. (2011) Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat Struct Mol Biol 18: 956–963.

61. KimTK, HembergM, GrayJM, CostaAM, BearDM, et al. (2010) Widespread transcription at neuronal activity-regulated enhancers. Nature 465: 182–187.

62. De SantaF, BarozziI, MiettonF, GhislettiS, PollettiS, et al. (2010) A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol 8: e1000384.

63. HeintzmanND, StuartRK, HonG, FuY, ChingCW, et al. (2007) Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet 39: 311–318.

64. DeisserothA, HendrickD (1978) Human alpha-globin gene expression following chromosomal dependent gene transfer into mouse erythroleukemia cells. Cell 15: 55–63.

65. DeisserothA, BurkR, PiccianoD, AndersonWF, NienhuisA, et al. (1975) Hemoglobin synthesis in somatic cell hybrids: globin gene expression in hybrids between mouse erythroleukemia and human marrow cells or fibroblasts. Proc Natl Acad Sci U S A 72: 1102–1106.

66. CraddockCF, VyasP, SharpeJA, AyyubH, WoodWG, et al. (1995) Contrasting effects of alpha and beta globin regulatory elements on chromatin structure may be related to their different chromosomal environments. EMBO J 14: 1718–1726.

67. JohnsonKD, GrassJA, ParkC, ImH, ChoiK, et al. (2003) Highly restricted localization of RNA polymerase II within a locus control region of a tissue-specific chromatin domain. Mol Cell Biol 23: 6484–6493.

68. YooEJ, CookeNE, LiebhaberSA (2012) An RNA-independent linkage of noncoding transcription to long-range enhancer function. Mol Cell Biol 32: 2020–2029.

69. WallaceHA, Marques-KrancF, RichardsonM, Luna-CrespoF, SharpeJA, et al. (2007) Manipulating the mouse genome to engineer precise functional syntenic replacements with human sequence. Cell 128: 197–209.

70. DevoyA, Bunton-StasyshynRK, TybulewiczVL, SmithAJ, FisherEM (2012) Genomically humanized mice: technologies and promises. Nat Rev Genet 13: 14–20.

71. VernimmenD, Marques-KrancF, SharpeJA, Sloane-StanleyJA, WoodWG, et al. (2009) Chromosome looping at the human alpha-globin locus is mediated via the major upstream regulatory element (HS -40). Blood 114: 4253–4260.

72. De GobbiM, AnguitaE, HughesJ, Sloane-StanleyJA, SharpeJA, et al. (2007) Tissue-specific histone modification and transcription factor binding in alpha globin gene expression. Blood 110: 4503–4510.

73. VernimmenD, LynchMD, De GobbiM, GarrickD, SharpeJA, et al. (2011) Polycomb eviction as a new distant enhancer function. Genes Dev 25: 1583–1588.

74. SawadoT, HalowJ, BenderMA, GroudineM (2003) The beta -globin locus control region (LCR) functions primarily by enhancing the transition from transcription initiation to elongation. Genes Dev 17: 1009–1018.

75. SongSH, KimA, RagoczyT, BenderMA, GroudineM, et al. (2010) Multiple functions of Ldb1 required for beta-globin activation during erythroid differentiation. Blood 116: 2356–2364.

76. Ghavi-HelmY, KleinFA, PakozdiT, CiglarL, NoordermeerD, et al. (2014) Enhancer loops appear stable during development and are associated with paused polymerase. Nature 512: 96–100.

77. BenderMA, RagoczyT, LeeJ, ByronR, TellingA, et al. (2012) The hypersensitive sites of the murine beta-globin locus control region act independently to affect nuclear localization and transcriptional elongation. Blood 119: 3820–3827.

78. StadhoudersR, ThongjueaS, Andrieu-SolerC, PalstraRJ, BryneJC, et al. (2012) Dynamic long-range chromatin interactions control Myb proto-oncogene transcription during erythroid development. EMBO J 31: 986–999.

79. LovenJ, HokeHA, LinCY, LauA, OrlandoDA, et al. (2013) Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153: 320–334.

80. ZentnerGE, TesarPJ, ScacheriPC (2011) Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res 21: 1273–1283.

81. HoY, ElefantF, LiebhaberSA, CookeNE (2006) Locus control region transcription plays an active role in long-range gene activation. Mol Cell 23: 365–375.

82. HoY, ElefantF, CookeN, LiebhaberS (2002) A defined locus control region determinant links chromatin domain acetylation with long-range gene activation. Mol Cell 9: 291–302.

83. ZhaoH, FriedmanRD, FournierRE (2007) The locus control region activates serpin gene expression through recruitment of liver-specific transcription factors and RNA polymerase II. Mol Cell Biol 27: 5286–5295.

84. SpicugliaS, KumarS, YehJH, VachezE, ChassonL, et al. (2002) Promoter activation by enhancer-dependent and -independent loading of activator and coactivator complexes. Mol Cell 10: 1479–1487.

85. TanimotoK, LiuQ, BungertJ, EngelJD (1999) Effects of altered gene order or orientation of the locus control region on human beta-globin gene expression in mice. Nature 398: 344–348.

86. BlackwoodEM, KadonagaJT (1998) Going the distance: a current view of enhancer action. Science 281: 60–63.

87. ZhuX, LingJ, ZhangL, PiW, WuM, et al. (2007) A facilitated tracking and transcription mechanism of long-range enhancer function. Nucleic Acids Res 35: 5532–5544.

88. HatzisP, TalianidisI (2002) Dynamics of enhancer-promoter communication during differentiation-induced gene activation. Mol Cell 10: 1467–1477.

89. WangQ, CarrollJS, BrownM (2005) Spatial and temporal recruitment of androgen receptor and its coactivators involves chromosomal looping and polymerase tracking. Mol Cell 19: 631–642.

90. TanM, LuoH, LeeS, JinF, YangJS, et al. (2011) Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell 146: 1016–1028.

91. SzutoriszH, DillonN, ToraL (2005) The role of enhancers as centres for general transcription factor recruitment. Trends Biochem Sci 30: 593–599.

92. StumpfM, YueX, SchmitzS, LucheH, ReddyJK, et al. (2010) Specific erythroid-lineage defect in mice conditionally deficient for Mediator subunit Med1. Proc Natl Acad Sci U S A 107: 21541–21546.

93. HiggsDR, VickersMA, WilkieAO, PretoriusIM, JarmanAP, et al. (1989) A review of the molecular genetics of the human alpha-globin gene cluster. Blood 73: 1081–1104.

94. VestriR, PieragostiniE, RistaldiMS (1994) Expression gradient in sheep alpha alpha and alpha alpha alpha globin gene haplotypes: mRNA levels. Blood 83: 2317–2322.

95. StamatoyannopoulosJA, SnyderM, HardisonR, RenB, GingerasT, et al. (2012) An encyclopedia of mouse DNA elements (Mouse ENCODE). Genome Biol 13: 418.

96. VieiraKF, LevingsPP, HillMA, CrusselleVJ, KangSH, et al. (2004) Recruitment of transcription complexes to the beta-globin gene locus in vivo and in vitro. J Biol Chem 279: 50350–50357.

97. GhamariA, van de CorputMP, ThongjueaS, van CappellenWA, van IjckenW, et al. (2013) In vivo live imaging of RNA polymerase II transcription factories in primary cells. Genes Dev 27: 767–777.

98. SutherlandH, BickmoreWA (2009) Transcription factories: gene expression in unions? Nat Rev Genet 10: 457–466.

99. WhyteWA, OrlandoDA, HniszD, AbrahamBJ, LinCY, et al. (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153: 307–319.

100. WendtKS, GrosveldFG (2014) Transcription in the context of the 3D nucleus. Curr Opin Genet Dev 25: 62–67.

101. IllingworthRS, BirdAP (2009) CpG islands–‘a rough guide’. FEBS Lett 583: 1713–1720.

102. SenR, GrosschedlR (2010) Memories of lost enhancers. Genes Dev 24: 973–979.

103. ChenS, MaJ, WuF, XiongLJ, MaH, et al. (2012) The histone H3 Lys 27 demethylase JMJD3 regulates gene expression by impacting transcriptional elongation. Genes Dev 26: 1364–1375.

104. WilliamsK, ChristensenJ, RappsilberJ, NielsenAL, JohansenJV, et al. (2014) The Histone Lysine Demethylase JMJD3/KDM6B Is Recruited to p53 Bound Promoters and Enhancer Elements in a p53 Dependent Manner. PLoS ONE 9: e96545.

105. TaberlayPC, KellyTK, LiuCC, YouJS, De CarvalhoDD, et al. (2011) Polycomb-repressed genes have permissive enhancers that initiate reprogramming. Cell 147: 1283–1294.

106. KondoT, IsonoK, KondoK, EndoTA, ItoharaS, et al. (2014) Polycomb potentiates meis2 activation in midbrain by mediating interaction of the promoter with a tissue-specific enhancer. Dev Cell 28: 94–101.

107. SeenundunS, RampalliS, LiuQC, AzizA, PaliiC, et al. (2010) UTX mediates demethylation of H3K27me3 at muscle-specific genes during myogenesis. EMBO J 29: 1401–1411.

108. OstuniR, PiccoloV, BarozziI, PollettiS, TermaniniA, et al. (2013) Latent enhancers activated by stimulation in differentiated cells. Cell 152: 157–171.

109. AndersonE, PelusoS, LetticeLA, HillRE (2012) Human limb abnormalities caused by disruption of hedgehog signaling. Trends Genet 28: 364–373.

110. SollainoMC, PagliettiME, LoiD, CongiuR, PoddaR, et al. (2010) Homozygous deletion of the major alpha-globin regulatory element (MCS-R2) responsible for a severe case of hemoglobin H disease. Blood 116: 2193–2194.

111. CoelhoA, PicancoI, SeuanesF, SeixasMT, FaustinoP (2010) Novel large deletions in the human alpha-globin gene cluster: Clarifying the HS-40 long-range regulatory role in the native chromosome environment. Blood Cells Mol Dis 45: 147–153.

112. LetticeLA, DanielsS, SweeneyE, VenkataramanS, DevenneyPS, et al. (2011) Enhancer-adoption as a mechanism of human developmental disease. Hum Mutat 32: 1492–1499.

113. LowerKM, De GobbiM, HughesJR, DerryCJ, AyyubH, et al. (2013) Analysis of sequence variation underlying tissue-specific transcription factor binding and gene expression. Hum Mutat 34: 1140–1148.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 10
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#