#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

TENET 2.0: Identification of key transcriptional regulators and enhancers in lung adenocarcinoma


Autoři: Daniel J. Mullen aff001;  Chunli Yan aff001;  Diane S. Kang aff001;  Beiyun Zhou aff003;  Zea Borok aff001;  Crystal N. Marconett aff001;  Peggy J. Farnham aff001;  Ite A. Offringa aff001;  Suhn Kyong Rhie aff001
Působiště autorů: Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America aff001;  Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America aff002;  Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, CA, United States of America aff003
Vyšlo v časopise: TENET 2.0: Identification of key transcriptional regulators and enhancers in lung adenocarcinoma. PLoS Genet 16(9): e32767. doi:10.1371/journal.pgen.1009023
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pgen.1009023

Souhrn

Lung cancer is the leading cause of cancer-related death and lung adenocarcinoma is its most common subtype. Although genetic alterations have been identified as drivers in subsets of lung adenocarcinoma, they do not fully explain tumor development. Epigenetic alterations have been implicated in the pathogenesis of tumors. To identify epigenetic alterations driving lung adenocarcinoma, we used an improved version of the Tracing Enhancer Networks using Epigenetic Traits method (TENET 2.0) in primary normal lung and lung adenocarcinoma cells. We found over 32,000 enhancers that appear differentially activated between normal lung and lung adenocarcinoma. Among the identified transcriptional regulators inactivated in lung adenocarcinoma vs. normal lung, NKX2-1 was linked to a large number of silenced enhancers. Among the activated transcriptional regulators identified, CENPA, FOXM1, and MYBL2 were linked to numerous cancer-specific enhancers. High expression of CENPA, FOXM1, and MYBL2 is particularly observed in a subgroup of lung adenocarcinomas and is associated with poor patient survival. Notably, CENPA, FOXM1, and MYBL2 are also key regulators of cancer-specific enhancers in breast adenocarcinoma of the basal subtype, but they are associated with distinct sets of activated enhancers. We identified individual lung adenocarcinoma enhancers linked to CENPA, FOXM1, or MYBL2 that were associated with poor patient survival. Knockdown experiments of FOXM1 and MYBL2 suggest that these factors regulate genes involved in controlling cell cycle progression and cell division. For example, we found that expression of TK1, a potential target gene of a MYBL2-linked enhancer, is associated with poor patient survival. Identification and characterization of key transcriptional regulators and associated enhancers in lung adenocarcinoma provides important insights into the deregulation of lung adenocarcinoma epigenomes, highlighting novel potential targets for clinical intervention.

Klíčová slova:

Adenocarcinoma of the lung – Breast cancer – DNA methylation – Gene expression – Lung and intrathoracic tumors – Regulator genes – Secondary lung tumors – Transcriptional control


Zdroje

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69: 7–34. doi: 10.3322/caac.21551

2. Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ CK (eds). SEER Cancer Statistics Review 1975–2016. 2019 p. Table 15.28. Available: https://seer.cancer.gov/csr/1975_2016/browse_csr.php?sectionSEL=15&pageSEL=sect_15_table.28

3. Malhotra J, Malvezzi M, Negri E, La Vecchia C, Boffetta P. Risk factors for lung cancer worldwide. Eur Respir J. 2016;48: 889–902. doi: 10.1183/13993003.00359–2016

4. Pesch B, Kendzia B, Gustavsson P, Jöckel KH, Johnen G, Pohlabeln H, et al. Cigarette smoking and lung cancer—Relative risk estimates for the major histological types from a pooled analysis of case—Control studies. Int J Cancer. 2012;131: 1210–1219. doi: 10.1002/ijc.27339

5. Cheng L, Alexander RE, MacLennan GT, Cummings OW, Montironi R, Lopez-Beltran A, et al. Molecular pathology of lung cancer: Key to personalized medicine. Mod Pathol. 2012;25: 347–369. doi: 10.1038/modpathol.2011.215

6. Zappa C, Mousa SA. Non-small cell lung cancer: Current treatment and future advances. Transl Lung Cancer Res. 2016;5: 288–300. doi: 10.21037/tlcr.2016.06.07

7. Collisson EA, Campbell JD, Brooks AN, Berger AH, Lee W, Chmielecki J, et al. Comprehensive molecular profiling of lung adenocarcinoma: The cancer genome atlas research network. Nature. 2014;511: 543–550. doi: 10.1038/nature13385

8. Roeder RG. The role of general initiation factors in transcription by RNA polymerase II. Trends Biochem Sci. 1996;21: 327–335. doi: 10.1016/0968-0004(96)10050-5

9. Stueve TR, Marconett CN, Zhou B, Borok Z, Laird-Offringa IA. The importance of detailed epigenomic profiling of different cell types within organs. Epigenomics. 2016;8: 817–829. doi: 10.2217/epi-2016-0005

10. Jones PA, Baylin SB. The Epigenomics of Cancer. Cell. 2007;128: 683–692. doi: 10.1016/j.cell.2007.01.029

11. Molina-Pinelo S. Epigenetics of lung cancer: a translational perspective. Cell Oncol. 2019. doi: 10.1007/s13402-019-00465-9

12. Rhie SK, Schreiner S, Witt H, Armoskus C, Lay FD, Camarena A, et al. Using 3D epigenomic maps of primary olfactory neuronal cells from living individuals to understand gene regulation. Sci Adv. 2018;4. doi: 10.1126/sciadv.aav8550

13. Yao L, Shen H, Laird PW, Farnham PJ, Berman BP. Inferring regulatory element landscapes and transcription factor networks from cancer methylomes. Genome Biol. 2015;16: 1–21. doi: 10.1186/s13059-015-0668-3

14. Rhie SK, Guo Y, Tak YG, Yao L, Shen H, Coetzee GA, et al. Identification of activated enhancers and linked transcription factors in breast, prostate, and kidney tumors by tracing enhancer networks using epigenetic traits. Epigenetics and Chromatin. 2016;9: 1–17. doi: 10.1186/s13072-016-0102-4

15. Xu J, Watts JA, Pope SD, Gadue P, Kamps M, Plath K, et al. Transcriptional competence and the active marking of tissue-specific enhancers by defined transcription factors in embryonic and induced pluripotent stem cells. Genes Dev. 2009;23: 2824–2838. doi: 10.1101/gad.1861209

16. Aran D, Sabato S, Hellman A. DNA methylation of distal regulatory sites characterizes dysregulation of cancer genes. Genome Biol. 2013;14. doi: 10.1186/gb-2013-14-3-r21

17. Silva TC, Coetzee SG, Gull N, Yao L, Hazelett DJ, Noushmehr H, et al. ELmer v.2: An r/bioconductor package to reconstruct gene regulatory networks from DNA methylation and transcriptome profiles. Bioinformatics. 2019;35: 1974–1977. doi: 10.1093/bioinformatics/bty902

18. Bulger M, Groudine M. Enhancers: The abundance and function of regulatory sequences beyond promoters. Dev Biol. 2010;339: 250–257. doi: 10.1016/j.ydbio.2009.11.035

19. Wright JC, Mudge J, Weisser H, Barzine MP, Gonzalez JM, Brazma A, et al. Improving GENCODE reference gene annotation using a high-stringency proteogenomics workflow. Nat Commun. 2016;7: 1–11. doi: 10.1038/ncomms11778

20. Lambert SA, Jolma A, Campitelli LF, Das PK, Yin Y, Albu M, et al. The Human Transcription Factors. Cell. 2018;172: 650–665. doi: 10.1016/j.cell.2018.01.029

21. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-small-cell lung cancers: A heterogeneous set of diseases. Nat Rev Cancer. 2014;14: 535–546. doi: 10.1038/nrc3775

22. Yang C, Stueve TR, Yan C, Rhie SK, Mullen DJ, Luo J, et al. Positional integration of lung adenocarcinoma susceptibility loci with primary human alveolar epithelial cell epigenomes. Epigenomics. 2018;10: 1167–1187. doi: 10.2217/epi-2018-0003

23. Marconett CN, Zhou B, Rieger ME, Selamat SA, Dubourd M, Fang X, et al. Integrated Transcriptomic and Epigenomic Analysis of Primary Human Lung Epithelial Cell Differentiation. PLoS Genet. 2013;9: 1–14. doi: 10.1371/journal.pgen.1003513

24. Roadmap Epigenomics Consortium, Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518: 317–329. doi: 10.1038/nature14248

25. Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489: 57–74. doi: 10.1038/nature11247

26. Davis CA, Hitz BC, Sloan CA, Chan ET, Davidson JM, Gabdank I, et al. The Encyclopedia of DNA elements (ENCODE): Data portal update. Nucleic Acids Res. 2018;46: D794–D801. doi: 10.1093/nar/gkx1081

27. Suzuki A, Kawano S, Mitsuyama T, Suyama M, Kanai Y, Shirahige K, et al. DBTSS/DBKERO for integrated analysis of transcriptional regulation. Nucleic Acids Res. 2018;46: D229–D238. doi: 10.1093/nar/gkx1001

28. Guler GD, Tindell CA, Pitti R, Wilson C, Nichols K, KaiWai Cheung T, et al. Repression of Stress-Induced LINE-1 Expression Protects Cancer Cell Subpopulations from Lethal Drug Exposure. Cancer Cell. 2017;32: 221–237.e13. doi: 10.1016/j.ccell.2017.07.002

29. Corces MR, Granja JM, Shams S, Louie BH, Seoane JA, Zhou W, et al. The chromatin accessibility landscape of primary human cancers. Science (80-). 2018;362: eaav1898. doi: 10.1126/science.aav1898

30. Wang Z, Tu K, Xia L, Luo K, Luo W, Tang J, et al. The open chromatin landscape of non–small cell lung carcinoma. Cancer Res. 2019;79: 4840–4854. doi: 10.1158/0008-5472.CAN-18-3663

31. Herriges M, Morrisey EE. Lung development: orchestrating the generation and regeneration of a complex organ. Development. 2014;141: 502–13. doi: 10.1242/dev.098186

32. Chen Y, Pacyna-Gengelbach M, Deutschmann N, Niesporek S, Petersen I. Homeobox gene HOP has a potential tumor suppressive activity in human lung cancer. Int J Cancer. 2007;121: 1021–1027. doi: 10.1002/ijc.22753

33. Rebouissou S, Vasiliu V, Thomas C, Bellanné-Chantelot C, Bui H, Chrétien Y, et al. Germline hepatocyte nuclear factor 1α and 1β mutations in renal cell carcinomas. Hum Mol Genet. 2005;14: 603–614. doi: 10.1093/hmg/ddi057

34. Terasawa K, Toyota M, Sagae S, Ogi K, Suzuki H, Sonoda T, et al. Epigenetic inactivation of TCF2 in ovarian cancer and various cancer cell lines. Br J Cancer. 2006;94: 914–921. doi: 10.1038/sj.bjc.6602984

35. Yu DD, Guo SW, Jing YY, Dong YL, Wei LX. A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci. 2015;5: 1–8. doi: 10.1186/s13578-015-0049-3

36. Gyorffy B, Surowiak P, Budczies J, Lánczky A. Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS One. 2013;8. doi: 10.1371/journal.pone.0082241

37. Wang I-C, Snyder J, Zhang Y, Lander J, Nakafuku Y, Lin J, et al. Foxm1 Mediates Cross Talk between Kras/Mitogen-Activated Protein Kinase and Canonical Wnt Pathways during Development of Respiratory Epithelium. Mol Cell Biol. 2012;32: 3838–3850. doi: 10.1128/mcb.00355-12

38. Hanada N, Lo HW, Day CP, Pan Y, Nakajima Y, Hung MC. Co-regulation of B-Myb expression by E2F1 and EGF receptor. Mol Carcinog. 2006;45: 10–17. doi: 10.1002/mc.20147

39. Rhie SK, Perez AA, Lay FD, Schreiner S, Shi J, Polin J, et al. A high-resolution 3D epigenomic map reveals insights into the creation of the prostate cancer transcriptome. Nat Commun. 2019;10: 4154. doi: 10.1038/s41467-019-12079-8

40. Wu Y, Du H, Zhan M, Wang H, Chen P, Du D, et al. Sepiapterin reductase promotes hepatocellular carcinoma progression via FoxO3a/Bim signaling in a nonenzymatic manner. Cell Death Dis. 2020;11. doi: 10.1038/s41419-020-2471-7

41. Winslow MM, Dayton TL, Verhaak RGW, Kim-Kiselak C, Snyder EL, Feldser DM, et al. Suppression of lung adenocarcinoma progression by Nkx2-1. Nature. 2011;473: 101–104. doi: 10.1038/nature09881

42. Rice SJ, Lai SC, Wood LW, Helsley KR, Runkle EA, Winslow MM, et al. MicroRNA-33a mediates the regulation of high mobility group AT-hook 2 gene (HMGA2) by thyroid transcription factor 1 (TTF-1/NKX2-1). J Biol Chem. 2013;288: 16348–16360. doi: 10.1074/jbc.M113.474643

43. Athwal RK, Walkiewicz MP, Baek S, Fu S, Bui M, Camps J, et al. CENP-A nucleosomes localize to transcription factor hotspots and subtelomeric sites in human cancer cells. Epigenetics and Chromatin. 2015;8: 1–23. doi: 10.1186/1756-8935-8-2

44. Sadasivam S, Duan S, DeCaprio JA. The MuvB complex sequentially recruits B-Myb and FoxM1 to promote mitotic gene expression. Genes Dev. 2012;26: 474–489. doi: 10.1101/gad.181933.111

45. Sanders DA, Gormally M V, Marsico G, Beraldi D, Tannahill D. FOXM1 binds directly to non-consensus sequences in the human genome. Genome Biol. 2015; 1–23. doi: 10.1186/s13059-015-0696-z

46. Sanders DA, Ross-Innes CS, Beraldi D, Carroll JS, Balasubramanian S. Genome-wide mapping of FOXM1 binding reveals co-binding with estrogen receptor alpha in breast cancer cells. Genome Biol. 2013;14: 1–16. doi: 10.1186/gb-2013-14-1-r6

47. Chae YK, Davis AA, Raparia K, Agte S, Pan A, Mohindra N, et al. Association of Tumor Mutational Burden With DNA Repair Mutations and Response to Anti–PD-1/PD-L1 Therapy in Non–Small-Cell Lung Cancer. Clin Lung Cancer. 2019;20: 88–96.e6. doi: 10.1016/j.cllc.2018.09.008

48. Zona S, Bella L, Burton MJ, Nestal de Moraes G, Lam EWF. FOXM1: An emerging master regulator of DNA damage response and genotoxic agent resistance. Biochim Biophys Acta—Gene Regul Mech. 2014;1839: 1316–1322. doi: 10.1016/j.bbagrm.2014.09.016

49. Rhie SK, Hazelett DJ, Coetzee SG, Yan C, Noushmehr H, Coetzee GA. Nucleosome positioning and histone modifications define relationships between regulatory elements and nearby gene expression in breast epithelial cells. BMC Genomics. 2014;15: 1–19. doi: 10.1186/1471-2164-15-331

50. Wang Y, Ung MH, Xia T, Cheng W, Cheng C. Cancer cell line specific co-factors modulate the FOXM1 cistrome. Oncotarget. 2017;8: 76498–76515. doi: 10.18632/oncotarget.20405

51. Wei P, Zhang N, Wang Y, Li D, Wang L, Sun X, et al. FOXM1 promotes lung adenocarcinoma invasion and metastasis by upregulating SNAIL. Int J Biol Sci. 2015;11: 186–198. doi: 10.7150/ijbs.10634

52. Zhang Y, Bin Qiao W, Shan L. Expression and functional characterization of FOXM1 in non-small cell lung cancer. Onco Targets Ther. 2018;11: 3385–3393. doi: 10.2147/OTT.S162523

53. Xiong YC, Wang J, Cheng Y, Zhang XY, Ye XQ. Overexpression of MYBL2 promotes proliferation and migration of non-small-cell lung cancer via upregulating NCAPH. Mol Cell Biochem. 2020;468: 185–193. doi: 10.1007/s11010-020-03721-x

54. Takeda DY, Spisák S, Seo JH, Bell C, O’Connor E, Korthauer K, et al. A Somatically Acquired Enhancer of the Androgen Receptor Is a Noncoding Driver in Advanced Prostate Cancer. Cell. 2018;174: 422–432.e13. doi: 10.1016/j.cell.2018.05.037

55. Corson TW, Huang A, Tsao MS, Gallie BL. KIF14 is a candidate oncogene in the 1q minimal region of genomic gain in multiple cancers. Oncogene. 2005;24: 4741–4753. doi: 10.1038/sj.onc.1208641

56. Iwakawa R, Kohno T, Kato M, Shiraishi K, Tsuta K, Noguchi M, et al. MYC amplification as a prognostic marker of early-stage lung adenocarcinoma identified by whole genome copy number analysis. Clin Cancer Res. 2011;17: 1481–1489. doi: 10.1158/1078-0432.CCR-10-2484

57. Mao S, Li Y, Lu Z, Che Y, Huang J, Lei Y, et al. PHD finger protein 5A promoted lung adenocarcinoma progression via alternative splicing. Cancer Med. 2019;8: 2429–2441. doi: 10.1002/cam4.2115

58. Morel M, Shah KN, Long W. The F-box protein FBXL16 up-regulates the stability of C-MYC oncoprotein by antagonizing the activity of the F-box protein FBW7. J Biol Chem. 2020;295: 7970–7980. doi: 10.1074/jbc.RA120.012658

59. Malvi P, Janostiak R, Nagarajan A, Cai G, Wajapeyee N. Loss of thymidine kinase 1 inhibits lung cancer growth and metastatic attributes by reducing GDF15 expression. PLOS Genet. 2019;15: e1008439. doi: 10.1371/journal.pgen.1008439

60. Uttarkar S, Frampton J, Klempnauer KH. Targeting the transcription factor Myb by small-molecule inhibitors. Exp Hematol. 2017;47: 31–35. doi: 10.1016/j.exphem.2016.12.003

61. Kwok JMM, Myatt SS, Marson CM, Coombes RC, Constantinidou D, Lam EWF. Thiostrepton selectively targets breast cancer cells through inhibition of forkhead box M1 expression. Mol Cancer Ther. 2008;7: 2022–2032. doi: 10.1158/1535-7163.MCT-08-0188

62. Gormally M V., Dexheimer TS, Marsico G, Sanders DA, Lowe C, Matak-Vinkoviä D, et al. Suppression of the FOXM1 transcriptional programme via novel small molecule inhibition. Nat Commun. 2014;5. doi: 10.1038/ncomms6165

63. Anders S, Pyl PT, Huber W. HTSeq-A Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31: 166–169. doi: 10.1093/bioinformatics/btu638

64. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15: 1–21. doi: 10.1186/s13059-014-0550-8

65. Zhu A, Ibrahim JG, Love MI. Heavy-Tailed prior distributions for sequence count data: Removing the noise and preserving large differences. Bioinformatics. 2019;35: 2084–2092. doi: 10.1093/bioinformatics/bty895

66. Mi H, Muruganujan A, Ebert D, Huang X, Thomas PD. PANTHER version 14: More genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 2019;47: D419–D426. doi: 10.1093/nar/gky1038

67. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009;462: 315–322. doi: 10.1038/nature08514

68. Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, et al. The NIH roadmap epigenomics mapping consortium. Nat Biotechnol. 2010;28: 1045–1048. doi: 10.1038/nbt1010-1045

69. Landt SG, Marinov GK, Kundaje A, Kheradpour P, Pauli F, Batzoglou S, et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 2012;22: 1813–1831. doi: 10.1101/gr.136184.111

70. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: A method for assaying chromatin accessibility genome-wide. Curr Protoc Mol Biol. 2015;2015: 21.29.1–21.29.9. doi: 10.1002/0471142727.mb2129s109

71. Koboldt DC, Fulton RS, McLellan MD, Schmidt H, Kalicki-Veizer J, McMichael JF, et al. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490: 61–70. doi: 10.1038/nature11412

72. Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. TCGAbiolinks: An R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44: e71. doi: 10.1093/nar/gkv1507

73. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Arman B, et al. In Focus The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. 2012. doi: 10.1158/2159-8290.CD-12-0095

74. Caligiuri MA, Dalton WS, Rodriguez L, Sellers T, Willman CL. Orien Reshaping Cancer Research & Treatment. Oncol Issues. 2016;31: 62–66. doi: 10.1080/10463356.2016.11884100

75. Dalton WS, Sullivan D, Ecsedy J, Caligiuri MA. Patient Enrichment for Precision-Based Cancer Clinical Trials: Using Prospective Cohort Surveillance as an Approach to Improve Clinical Trials. Clin Pharmacol Ther. 2018;104: 23–26. doi: 10.1002/cpt.1051

76. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6: 1–20. doi: 10.1126/scisignal.2004088

77. Kulakovskiy I V., Vorontsov IE, Yevshin IS, Sharipov RN, Fedorova AD, Rumynskiy EI, et al. HOCOMOCO: Towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-Seq analysis. Nucleic Acids Res. 2018;46: D252–D259. doi: 10.1093/nar/gkx1106

78. Grant CE, Bailey TL, Noble WS. FIMO: Scanning for occurrences of a given motif. Bioinformatics. 2011;27: 1017–1018. doi: 10.1093/bioinformatics/btr064

79. Rao SSP, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. 2014;159: 1665–1680. doi: 10.1016/j.cell.2014.11.021

80. Wang Y, Song F, Zhang B, Zhang L, Xu J, Kuang D, et al. The 3D Genome Browser: A web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol. 2018;19: 1–12. doi: 10.1186/s13059-018-1519-9


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 9
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#