#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Integrative Analysis of DNA Methylation and Gene Expression Data Identifies as a Key Regulator of COPD


Chronic Obstructive Pulmonary Disease (COPD) is a common lung disease. It is the fourth leading cause of death in the world and is expected to be the third by 2020. COPD is a heterogeneous and complex disease consisting of obstruction in the small airways, emphysema, and chronic bronchitis. COPD is generally caused by exposure to noxious particles or gases, most commonly from cigarette smoking. However, only 20–25% of smokers develop clinically significant airflow obstruction. Smoking is known to cause epigenetic changes in lung tissues. Thus, genetics, epigenetic, and their interaction with environmental factors play an important role in COPD pathogenesis and progression. Currently, there are no therapeutics that can reverse COPD progression. In order to identify new targets that may lead to the development of therapeutics for curing COPD, we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity.


Vyšlo v časopise: Integrative Analysis of DNA Methylation and Gene Expression Data Identifies as a Key Regulator of COPD. PLoS Genet 11(1): e32767. doi:10.1371/journal.pgen.1004898
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004898

Souhrn

Chronic Obstructive Pulmonary Disease (COPD) is a common lung disease. It is the fourth leading cause of death in the world and is expected to be the third by 2020. COPD is a heterogeneous and complex disease consisting of obstruction in the small airways, emphysema, and chronic bronchitis. COPD is generally caused by exposure to noxious particles or gases, most commonly from cigarette smoking. However, only 20–25% of smokers develop clinically significant airflow obstruction. Smoking is known to cause epigenetic changes in lung tissues. Thus, genetics, epigenetic, and their interaction with environmental factors play an important role in COPD pathogenesis and progression. Currently, there are no therapeutics that can reverse COPD progression. In order to identify new targets that may lead to the development of therapeutics for curing COPD, we developed a systematic approach to identify key regulators of COPD that integrates genome-wide DNA methylation, gene expression, and phenotype data in lung tissue from COPD and control samples. Our integrative analysis identified 126 key regulators of COPD. We identified EPAS1 as the only key regulator whose downstream genes significantly overlapped with multiple genes sets associated with COPD disease severity.


Zdroje

1. LopezAD, ShibuyaK, RaoC, MathersCD, HansellAL, et al. (2006) Chronic obstructive pulmonary disease: current burden and future projections. Eur Respir J 27: 397–412.

2. AgustiA, CalverleyPM, CelliB, CoxsonHO, EdwardsLD, et al. (2010) Characterisation of COPD heterogeneity in the ECLIPSE cohort. Respir Res 11: 122.

3. RabeKF, HurdS, AnzuetoA, BarnesPJ, BuistSA, et al. (2007) Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med 176: 532–555.

4. HarveyBG, HeguyA, LeopoldPL, CarolanBJ, FerrisB, et al. (2007) Modification of gene expression of the small airway epithelium in response to cigarette smoking. J Mol Med (Berl) 85: 39–53.

5. Sethi JM, Rochester CL (2000) Smoking and chronic obstructive pulmonary disease. Clin Chest Med 21: 67–86, viii.

6. ShapiroSD, IngenitoEP (2005) The pathogenesis of chronic obstructive pulmonary disease: advances in the past 100 years. Am J Respir Cell Mol Biol 32: 367–372.

7. LokkeA, LangeP, ScharlingH, FabriciusP, VestboJ (2006) Developing COPD: a 25 year follow up study of the general population. Thorax 61: 935–939.

8. MayerAS, NewmanLS (2001) Genetic and environmental modulation of chronic obstructive pulmonary disease. Respir Physiol 128: 3–11.

9. KentL, SmythL, ClaytonC, ScottL, CookT, et al. (2008) Cigarette smoke extract induced cytokine and chemokine gene expression changes in COPD macrophages. Cytokine 42: 205–216.

10. NingW, LiCJ, KaminskiN, Feghali-BostwickCA, AlberSM, et al. (2004) Comprehensive gene expression profiles reveal pathways related to the pathogenesis of chronic obstructive pulmonary disease. Proc Natl Acad Sci U S A 101: 14895–14900.

11. BosseY, PostmaDS, SinDD, LamontagneM, CoutureC, et al. (2012) Molecular signature of smoking in human lung tissues. Cancer Res 72: 3753–3763.

12. ChoMH, CastaldiPJ, WanES, SiedlinskiM, HershCP, et al. (2012) A genome-wide association study of COPD identifies a susceptibility locus on chromosome 19q13. Hum Mol Genet 21: 947–957.

13. PillaiSG, GeD, ZhuG, KongX, ShiannaKV, et al. (2009) A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet 5: e1000421.

14. EggerG, LiangG, AparicioA, JonesPA (2004) Epigenetics in human disease and prospects for epigenetic therapy. Nature 429: 457–463.

15. LepeuleJ, BaccarelliA, MottaV, CantoneL, LitonjuaAA, et al. (2012) Gene promoter methylation is associated with lung function in the elderly: the Normative Aging Study. Epigenetics 7: 261–269.

16. SelamatSA, ChungBS, GirardL, ZhangW, ZhangY, et al. (2012) Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res 22: 1197–1211.

17. Kalari S, Jung M, Kernstine KH, Takahashi T, Pfeifer GP (2012) The DNA methylation landscape of small cell lung cancer suggests a differentiation defect of neuroendocrine cells. Oncogene.

18. AdcockIM, FordP, ItoK, BarnesPJ (2006) Epigenetics and airways disease. Respir Res 7: 21.

19. LeeKW, PausovaZ (2013) Cigarette smoking and DNA methylation. Front Genet 4: 132.

20. BreitlingLP, YangR, KornB, BurwinkelB, BrennerH (2011) Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet 88: 450–457.

21. TanQ, WangG, HuangJ, DingZ, LuoQ, et al. (2013) Epigenomic analysis of lung adenocarcinoma reveals novel DNA methylation patterns associated with smoking. Onco Targets Ther 6: 1471–1479.

22. Buro-AuriemmaLJ, SalitJ, HackettNR, WaltersMS, Strulovici-BarelY, et al. (2013) Cigarette smoking induces small airway epithelial epigenetic changes with corresponding modulation of gene expression. Hum Mol Genet 22: 4726–4738.

23. QiuW, BaccarelliA, CareyVJ, BoutaouiN, BachermanH, et al. (2012) Variable DNA methylation is associated with chronic obstructive pulmonary disease and lung function. Am J Respir Crit Care Med 185: 373–381.

24. VucicEA, ChariR, ThuKL, WilsonIM, CottonAM, et al. (2014) DNA methylation is globally disrupted and associated with expression changes in chronic obstructive pulmonary disease small airways. Am J Respir Cell Mol Biol 50: 912–922.

25. YooS, HuangT, CampbellJD, LeeE, TuZ, et al. (2014) MODMatcher: Multi-Omics Data Matcher for Integrative Genomic Analysis. PLoS Comput Biol 10: e1003790.

26. SiegmundKD (2011) Statistical approaches for the analysis of DNA methylation microarray data. Hum Genet 129: 585–595.

27. AryeeMJ, WuZ, Ladd-AcostaC, HerbB, FeinbergAP, et al. (2011) Accurate genome-scale percentage DNA methylation estimates from microarray data. Biostatistics 12: 197–210.

28. WuH, CaffoB, JaffeeHA, IrizarryRA, FeinbergAP (2010) Redefining CpG islands using hidden Markov models. Biostatistics 11: 499–514.

29. BrenetF, MohM, FunkP, FeiersteinE, VialeAJ, et al. (2011) DNA methylation of the first exon is tightly linked to transcriptional silencing. PLoS One 6: e14524.

30. DoiA, ParkIH, WenB, MurakamiP, AryeeMJ, et al. (2009) Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet 41: 1350–1353.

31. HanH, CortezCC, YangX, NicholsPW, JonesPA, et al. (2011) DNA methylation directly silences genes with non-CpG island promoters and establishes a nucleosome occupied promoter. Hum Mol Genet 20: 4299–4310.

32. Bahar HalpernK, VanaT, WalkerMD (2014) Paradoxical Role of DNA Methylation in Activation of FoxA2 Gene Expression during Endoderm Development. J Biol Chem 289: 23882–23892.

33. NiesenMI, OsborneAR, YangH, RastogiS, ChellappanS, et al. (2005) Activation of a methylated promoter mediated by a sequence-specific DNA-binding protein, RFX. J Biol Chem 280: 38914–38922.

34. PeiferM, Fernandez-CuestaL, SosML, GeorgeJ, SeidelD, et al. (2012) Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet 44: 1104–1110.

35. StankiewiczP, SenP, BhattSS, StorerM, XiaZ, et al. (2009) Genomic and genic deletions of the FOX gene cluster on 16q24.1 and inactivating mutations of FOXF1 cause alveolar capillary dysplasia and other malformations. Am J Hum Genet 84: 780–791.

36. WanH, DingleS, XuY, BesnardV, KaestnerKH, et al. (2005) Compensatory roles of Foxa1 and Foxa2 during lung morphogenesis. J Biol Chem 280: 13809–13816.

37. YangF, TangX, RiquelmeE, BehrensC, NilssonMB, et al. (2011) Increased VEGFR-2 gene copy is associated with chemoresistance and shorter survival in patients with non-small-cell lung carcinoma who receive adjuvant chemotherapy. Cancer Res 71: 5512–5521.

38. BrockMV, HookerCM, Ota-MachidaE, HanY, GuoM, et al. (2008) DNA methylation markers and early recurrence in stage I lung cancer. N Engl J Med 358: 1118–1128.

39. YangM, ParkJY (2012) DNA methylation in promoter region as biomarkers in prostate cancer. Methods Mol Biol 863: 67–109.

40. BisharaAJ, HittnerJB (2012) Testing the Significance of a Correlation With Nonnormal Data: Comparison of Pearson, Spearman, Transformation, and Resampling Approaches. Psychological Methods 17: 399–417.

41. HolmesR, SolowayPD (2006) Regulation of imprinted DNA methylation. Cytogenetic and Genome Research 113: 122–129.

42. BellJT, PaiAA, PickrellJK, GaffneyDJ, Pique-RegiR, et al. (2011) DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol 12: R10.

43. LuoJ, YuY, ZhangH, TianF, ChangS, et al. (2011) Down-regulation of promoter methylation level of CD4 gene after MDV infection in MD-susceptible chicken line. BMC Proc 5 Suppl 4S7.

44. WangY, WysockaJ, SayeghJ, LeeYH, PerlinJR, et al. (2004) Human PAD4 regulates histone arginine methylation levels via demethylimination. Science 306: 279–283.

45. SchadtEE, LambJ, YangX, ZhuJ, EdwardsS, et al. (2005) An integrative genomics approach to infer causal associations between gene expression and disease. Nat Genet 37: 710–717.

46. ChenLS, Emmert-StreibF, StoreyJD (2007) Harnessing naturally randomized transcription to infer regulatory relationships among genes. Genome Biol 8: R219.

47. MillsteinJ, ZhangB, ZhuJ, SchadtEE (2009) Disentangling molecular relationships with a causal inference test. BMC Genet 10: 23.

48. MacintyreN, CrapoRO, ViegiG, JohnsonDC, van der GrintenCP, et al. (2005) Standardisation of the single-breath determination of carbon monoxide uptake in the lung. Eur Respir J 26: 720–735.

49. CelliBR, CoteCG, LareauSC, MeekPM (2008) Predictors of Survival in COPD: more than just the FEV1. Respir Med 102 Suppl 1S27–35.

50. PauwelsRA, BuistAS, CalverleyPM, JenkinsCR, HurdSS (2001) Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir Crit Care Med 163: 1256–1276.

51. CampbellJD, McDonoughJE, ZeskindJE, HackettTL, PechkovskyDV, et al. (2012) A gene expression signature of emphysema-related lung destruction and its reversal by the tripeptide GHK. Genome Med 4: 67.

52. HoggJC, ChuF, UtokaparchS, WoodsR, ElliottWM, et al. (2004) The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350: 2645–2653.

53. RenzoniEA, AbrahamDJ, HowatS, Shi-WenX, SestiniP, et al. (2004) Gene expression profiling reveals novel TGFbeta targets in adult lung fibroblasts. Respir Res 5: 24.

54. TianH, McKnightSL, RussellDW (1997) Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev 11: 72–82.

55. EmaM, TayaS, YokotaniN, SogawaK, MatsudaY, et al. (1997) A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc Natl Acad Sci U S A 94: 4273–4278.

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

57. 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.

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

59. PouyssegurJ, DayanF, MazureNM (2006) Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441: 437–443.

60. CovelloKL, KehlerJ, YuH, GordanJD, ArshamAM, et al. (2006) HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev 20: 557–570.

61. CovelloKL, SimonMC, KeithB (2005) Targeted replacement of hypoxia-inducible factor-1alpha by a hypoxia-inducible factor-2alpha knock-in allele promotes tumor growth. Cancer Res 65: 2277–2286.

62. SkuliN, LiuL, RungeA, WangT, YuanL, et al. (2009) Endothelial deletion of hypoxia-inducible factor-2alpha (HIF-2alpha) alters vascular function and tumor angiogenesis. Blood 114: 469–477.

63. ManaloDJ, RowanA, LavoieT, NatarajanL, KellyBD, et al. (2005) Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood 105: 659–669.

64. ElvidgeGP, GlennyL, AppelhoffRJ, RatcliffePJ, RagoussisJ, et al. (2006) Concordant regulation of gene expression by hypoxia and 2-oxoglutarate-dependent dioxygenase inhibition: the role of HIF-1alpha, HIF-2alpha, and other pathways. J Biol Chem 281: 15215–15226.

65. JiangY, ZhangW, KondoK, KlcoJM, St MartinTB, et al. (2003) Gene expression profiling in a renal cell carcinoma cell line: dissecting VHL and hypoxia-dependent pathways. Mol Cancer Res 1: 453–462.

66. WeinmannM, BelkaC, GunerD, GoeckeB, MullerI, et al. (2005) Array-based comparative gene expression analysis of tumor cells with increased apoptosis resistance after hypoxic selection. Oncogene 24: 5914–5922.

67. WinterSC, BuffaFM, SilvaP, MillerC, ValentineHR, et al. (2007) Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res 67: 3441–3449.

68. GuerassimovA, HoshinoY, TakuboY, TurcotteA, YamamotoM, et al. (2004) The development of emphysema in cigarette smoke-exposed mice is strain dependent. Am J Respir Crit Care Med 170: 974–980.

69. ForonjyRF, MercerBA, MaxfieldMW, PowellCA, D'ArmientoJ, et al. (2005) Structural emphysema does not correlate with lung compliance: lessons from the mouse smoking model. Exp Lung Res 31: 547–562.

70. MirrakhimovAE (2012) Chronic obstructive pulmonary disease and glucose metabolism: a bitter sweet symphony. Cardiovasc Diabetol 11: 132.

71. ZhangL, GjoerupO, RobertsTM (2004) The serine/threonine kinase cyclin G-associated kinase regulates epidermal growth factor receptor signaling. Proc Natl Acad Sci U S A 101: 10296–10301.

72. TabaraH, NaitoY, ItoA, KatsumaA, SakuraiMA, et al. (2011) Neonatal lethality in knockout mice expressing the kinase-dead form of the gefitinib target GAK is caused by pulmonary dysfunction. PLoS One 6: e26034.

73. BowlerRP (2012) Surfactant protein D as a biomarker for chronic obstructive pulmonary disease. COPD 9: 651–653.

74. KentBD, MitchellPD, McNicholasWT (2011) Hypoxemia in patients with COPD: cause, effects, and disease progression. Int J Chron Obstruct Pulmon Dis 6: 199–208.

75. UchidaT, RossignolF, MatthayMA, MounierR, CouetteS, et al. (2004) Prolonged hypoxia differentially regulates hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha expression in lung epithelial cells: implication of natural antisense HIF-1alpha. J Biol Chem 279: 14871–14878.

76. FormentiF, BeerPA, CroftQP, DorringtonKL, GaleDP, et al. (2011) Cardiopulmonary function in two human disorders of the hypoxia-inducible factor (HIF) pathway: von Hippel-Lindau disease and HIF-2alpha gain-of-function mutation. FASEB J 25: 2001–2011.

77. PengYJ, NanduriJ, KhanSA, YuanG, WangN, et al. (2011) Hypoxia-inducible factor 2alpha (HIF-2alpha) heterozygous-null mice exhibit exaggerated carotid body sensitivity to hypoxia, breathing instability, and hypertension. Proc Natl Acad Sci U S A 108: 3065–3070.

78. BrusselmansK, CompernolleV, TjwaM, WiesenerMS, MaxwellPH, et al. (2003) Heterozygous deficiency of hypoxia-inducible factor-2alpha protects mice against pulmonary hypertension and right ventricular dysfunction during prolonged hypoxia. J Clin Invest 111: 1519–1527.

79. VoelkelNF, VandivierRW, TuderRM (2006) Vascular endothelial growth factor in the lung. Am J Physiol Lung Cell Mol Physiol 290: L209–221.

80. CompernolleV, BrusselmansK, AckerT, HoetP, TjwaM, et al. (2002) Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nat Med 8: 702–710.

81. SinDD, PahlavanPS, ManSF (2008) Surfactant protein D: a lung specific biomarker in COPD? Ther Adv Respir Dis 2: 65–74.

82. CastaldiPJ, ChoMH, CohnM, LangermanF, MoranS, et al. (2010) The COPD genetic association compendium: a comprehensive online database of COPD genetic associations. Hum Mol Genet 19: 526–534.

83. ChanockSJ, ManolioT, BoehnkeM, BoerwinkleE, HunterDJ, et al. (2007) Replicating genotype-phenotype associations. Nature 447: 655–660.

84. GingoMR, SilveiraLJ, MillerYE, FriedlanderAL, CosgroveGP, et al. (2008) Tumour necrosis factor gene polymorphisms are associated with COPD. Eur Respir J 31: 1005–1012.

85. BosseY (2012) Updates on the COPD gene list. Int J Chron Obstruct Pulmon Dis 7: 607–631.

86. Vucic EA, Chari R, Thu KL, Wilson IM, Cotton AM, et al. (2013) DNA Methylation is Globally Disrupted and Associated with Expression Changes in COPD Small Airways. Am J Respir Cell Mol Biol.

87. RobinsonCM, NearyR, LevendaleA, WatsonCJ, BaughJA (2012) Hypoxia-induced DNA hypermethylation in human pulmonary fibroblasts is associated with Thy-1 promoter methylation and the development of a pro-fibrotic phenotype. Respir Res 13: 74.

88. WatsonJA, WatsonCJ, McCrohanAM, WoodfineK, TosettoM, et al. (2009) Generation of an epigenetic signature by chronic hypoxia in prostate cells. Hum Mol Genet 18: 3594–3604.

89. FimognariFL, LoffredoL, Di SimoneS, SampietroF, PastorelliR, et al. (2009) Hyperhomocysteinaemia and poor vitamin B status in chronic obstructive pulmonary disease. Nutr Metab Cardiovasc Dis 19: 654–659.

90. ReiterV, MatschkalDM, WagnerM, GlobischD, KneuttingerAC, et al. (2012) The CDK5 repressor CDK5RAP1 is a methylthiotransferase acting on nuclear and mitochondrial RNA. Nucleic Acids Res 40: 6235–6240.

91. KulpDC, JagalurM (2006) Causal inference of regulator-target pairs by gene mapping of expression phenotypes. BMC Genomics 7: 125.

92. YangX, DeignanJL, QiH, ZhuJ, QianS, et al. (2009) Validation of candidate causal genes for obesity that affect shared metabolic pathways and networks. Nat Genet 41: 415–423.

93. KinoseD, OgawaE, HirotaT, ItoI, KudoM, et al. (2012) A NOD2 gene polymorphism is associated with the prevalence and severity of chronic obstructive pulmonary disease in a Japanese population. Respirology 17: 164–171.

94. GopalP, ReynaertNL, ScheijenJL, SchalkwijkCG, FranssenFM, et al. (2014) Association of plasma sRAGE, but not esRAGE with lung function impairment in COPD. Respir Res 15: 24.

95. NanceT, SmithKS, AnayaV, RichardsonR, HoL, et al. (2014) Transcriptome analysis reveals differential splicing events in IPF lung tissue. PLoS One 9: e97550.

96. TahilianiM, KohKP, ShenY, PastorWA, BandukwalaH, et al. (2009) Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324: 930–935.

97. SzulwachKE, LiX, LiY, SongCX, WuH, et al. (2011) 5-hmC-mediated epigenetic dynamics during postnatal neurodevelopment and aging. Nat Neurosci 14: 1607–1616.

98. KriaucionisS, HeintzN (2009) The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324: 929–930.

99. GlobischD, MunzelM, MullerM, MichalakisS, WagnerM, et al. (2010) Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PLoS One 5: e15367.

100. IrizarryRA, Ladd-AcostaC, CarvalhoB, WuH, BrandenburgSA, et al. (2008) Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res 18: 780–790.

101. AshburnerM, BallCA, BlakeJA, BotsteinD, ButlerH, et al. (2000) Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25: 25–29.

102. FalconS, GentlemanR (2007) Using GOstats to test gene lists for GO term association. Bioinformatics 23: 257–258.

103. TrapnellC, RobertsA, GoffL, PerteaG, KimD, et al. (2012) Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7: 562–578.

104. StoreyJD, TibshiraniR (2003) Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 100: 9440–9445.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 1
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#