#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Identification of Chemical Inhibitors of β-Catenin-Driven Liver Tumorigenesis in Zebrafish


Liver cancer is a leading cause of cancer-related death. Genetic analysis of liver cancer has enabled classification of these tumors into subsets with unique genetic, clinical, and prognostic features. The search for targeted liver cancer treatments has been hampered by the lack of relevant animal models for these genetically diverse subsets, including liver cancers that are defined by activating mutations in the gene encoding β-catenin, an integral component of the Wnt signaling pathway. Here we describe the generation and characterization of genetically modified zebrafish expressing hepatocyte-specific activated β-catenin. We used this new zebrafish model to screen for drugs that suppress β-catenin-induced liver growth, and identified two classes of hits, c-Jun N-terminal kinase (JNK) inhibitors and antidepressants, that suppressed this phenotype. Our findings provide insights into the mechanisms by which β-catenin promotes liver tumor formation and implicate JNK inhibitors and antidepressants as potential treatments for a subset of human liver cancers.


Vyšlo v časopise: Identification of Chemical Inhibitors of β-Catenin-Driven Liver Tumorigenesis in Zebrafish. PLoS Genet 11(7): e32767. doi:10.1371/journal.pgen.1005305
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005305

Souhrn

Liver cancer is a leading cause of cancer-related death. Genetic analysis of liver cancer has enabled classification of these tumors into subsets with unique genetic, clinical, and prognostic features. The search for targeted liver cancer treatments has been hampered by the lack of relevant animal models for these genetically diverse subsets, including liver cancers that are defined by activating mutations in the gene encoding β-catenin, an integral component of the Wnt signaling pathway. Here we describe the generation and characterization of genetically modified zebrafish expressing hepatocyte-specific activated β-catenin. We used this new zebrafish model to screen for drugs that suppress β-catenin-induced liver growth, and identified two classes of hits, c-Jun N-terminal kinase (JNK) inhibitors and antidepressants, that suppressed this phenotype. Our findings provide insights into the mechanisms by which β-catenin promotes liver tumor formation and implicate JNK inhibitors and antidepressants as potential treatments for a subset of human liver cancers.


Zdroje

1. Mínguez B, Tovar V, Chiang D, Villanueva A, Llovet JM. Pathogenesis of hepatocellular carcinoma and molecular therapies. Curr Opin Gastroenterol. 2009;25: 186–194. doi: 10.1097/MOG.0b013e32832962a1 19387255

2. Wrzesinski SH, Taddei TH, Strazzabosco M. Systemic therapy in hepatocellular carcinoma. Clin Liver Dis. 2011;15: 423–441, vii–x. doi: 10.1016/j.cld.2011.03.002 21689622

3. Chiang DY, Villanueva A, Hoshida Y, Peix J, Newell P, Minguez B, et al. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68: 6779–6788. doi: 10.1158/0008-5472.CAN-08-0742 18701503

4. Laurent-Puig P, Legoix P, Bluteau O, Belghiti J, Franco D, Binot F, et al. Genetic alterations associated with hepatocellular carcinomas define distinct pathways of hepatocarcinogenesis. Gastroenterology. 2001;120: 1763–1773. 11375957

5. Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatol. 2007;45: 42–52. doi: 10.1002/hep.21467

6. Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003;1653: 1–24. 12781368

7. Zucman-Rossi J, Jeannot E, Nhieu JTV, Scoazec J- Y, Guettier C, Rebouissou S, et al. Genotype-phenotype correlation in hepatocellular adenoma: new classification and relationship with HCC. Hepatol. 2006;43: 515–524. doi: 10.1002/hep.21068

8. Pilati C, Letouzé E, Nault J-C, Imbeaud S, Boulai A, Calderaro J, et al. Genomic profiling of hepatocellular adenomas reveals recurrent FRK-activating mutations and the mechanisms of malignant transformation. Cancer Cell. 2014;25: 428–441. doi: 10.1016/j.ccr.2014.03.005 24735922

9. Colnot S, Decaens T, Niwa-Kawakita M, Godard C, Hamard G, Kahn A, et al. Liver-targeted disruption of Apc in mice activates beta-catenin signaling and leads to hepatocellular carcinomas. Proc Natl Acad Sci U S A. 2004;101: 17216–17221. doi: 10.1073/pnas.0404761101 15563600

10. Haramis A-PG, Hurlstone A, van der Velden Y, Begthel H, van den Born M, Offerhaus GJA, et al. Adenomatous polyposis coli-deficient zebrafish are susceptible to digestive tract neoplasia. EMBO Rep. 2006;7: 444–449. doi: 10.1038/sj.embor.7400638 16439994

11. Nejak-Bowen KN, Thompson MD, Singh S, Bowen WC, Dar MJ, Khillan J, et al. Accelerated liver regeneration and hepatocarcinogenesis in mice overexpressing serine-45 mutant beta-catenin. Hepatol. 2010;51: 1603–1613. doi: 10.1002/hep.23538

12. Harada N, Oshima H, Katoh M, Tamai Y, Oshima M, Taketo MM. Hepatocarcinogenesis in mice with beta-catenin and Ha-ras gene mutations. Cancer Res. 2004;64: 48–54. 14729607

13. Nejak-Bowen KN, Monga SPS. Beta-catenin signaling, liver regeneration and hepatocellular cancer: sorting the good from the bad. Semin Cancer Biol. 2011;21: 44–58. doi: 10.1016/j.semcancer.2010.12.010 21182948

14. Zon LI, Peterson RT. In vivo drug discovery in the zebrafish. Nat Rev Drug Discov. 2005;4: 35–44. doi: 10.1038/nrd1606 15688071

15. Liu S, Leach SD. Zebrafish models for cancer. Annu Rev Pathol. 2011;6: 71–93. doi: 10.1146/annurev-pathol-011110-130330 21261518

16. Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT. The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 1996;10: 1443–1454. 8666229

17. Her GM, Yeh Y-H, Wu J-L. 435-bp liver regulatory sequence in the liver fatty acid binding protein (L-FABP) gene is sufficient to modulate liver regional expression in transgenic zebrafish. Dev Dyn. 2003;227: 347–356. doi: 10.1002/dvdy.10324 12815620

18. Miyoshi Y, Iwao K, Nagasawa Y, Aihara T, Sasaki Y, Imaoka S, et al. Activation of the beta-catenin gene in primary hepatocellular carcinomas by somatic alterations involving exon 3. Cancer Res. 1998;58: 2524–2527. 9635572

19. Kurita R, Sagara H, Aoki Y, Link BA, Arai K, Watanabe S. Suppression of lens growth by alphaA-crystallin promoter-driven expression of diphtheria toxin results in disruption of retinal cell organization in zebrafish. Dev Biol. 2003;255: 113–127. 12618137

20. Hesselson D, Anderson RM, Beinat M, Stainier DYR. Distinct populations of quiescent and proliferative pancreatic beta-cells identified by HOTcre mediated labeling. Proc Natl Acad Sci U S A. 2009;106: 14896–14901. doi: 10.1073/pnas.0906348106 19706417

21. Moro E, Ozhan-Kizil G, Mongera A, Beis D, Wierzbicki C, Young RM, et al. In vivo Wnt signaling tracing through a transgenic biosensor fish reveals novel activity domains. Dev Biol. 2012;366: 327–340. doi: 10.1016/j.ydbio.2012.03.023 22546689

22. Mudbhary R, Hoshida Y, Chernyavskaya Y, Jacob V, Villanueva A, Fiel MI, et al. UHRF1 overexpression drives DNA hypomethylation and hepatocellular carcinoma. Cancer Cell. 2014;25: 196–209. doi: 10.1016/j.ccr.2014.01.003 24486181

23. Kundu R, Handa U, Mohan H. Role of DNA flow cytometry and immunocytochemical analysis in diagnosis of malignant effusions. Diagn Cytopathol. 2012;40: 887–892. doi: 10.1002/dc.21673 21485027

24. Berghmans S, Murphey RD, Wienholds E, Neuberg D, Kutok JL, Fletcher CDM, et al. tp53 mutant zebrafish develop malignant peripheral nerve sheath tumors. Proc Natl Acad Sci U S A. 2005;102: 407–412. doi: 10.1073/pnas.0406252102 15630097

25. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30: 207–210. 11752295

26. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281: 1509–1512. 9727977

27. Hovanes K, Li TW, Munguia JE, Truong T, Milovanovic T, Lawrence Marsh J, et al. Beta-catenin-sensitive isoforms of lymphoid enhancer factor-1 are selectively expressed in colon cancer. Nat Genet. 2001;28: 53–57. doi: 10.1038/88264 11326276

28. He TC, Chan TA, Vogelstein B, Kinzler KW. PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell. 1999;99: 335–345. 10555149

29. Weidinger G, Thorpe CJ, Wuennenberg-Stapleton K, Ngai J, Moon RT. The Sp1-related transcription factors sp5 and sp5-like act downstream of Wnt/beta-catenin signaling in mesoderm and neuroectoderm patterning. Curr Biol. 2005;15: 489–500. doi: 10.1016/j.cub.2005.01.041 15797017

30. Tung EK-K, Mak CK-M, Fatima S, Lo RC-L, Zhao H, Zhang C, et al. Clinicopathological and prognostic significance of serum and tissue Dickkopf-1 levels in human hepatocellular carcinoma. Liver Int. 2011;31: 1494–1504. doi: 10.1111/j.1478-3231.2011.02597.x 21955977

31. Lamb JR, Zhang C, Xie T, Wang K, Zhang B, Hao K, et al. Predictive genes in adjacent normal tissue are preferentially altered by sCNV during tumorigenesis in liver cancer and may rate limiting. PloS One. 2011;6: e20090. doi: 10.1371/journal.pone.0020090 21750698

32. Sung W-K, Zheng H, Li S, Chen R, Liu X, Li Y, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 2012;44: 765–769. doi: 10.1038/ng.2295 22634754

33. Bennett BL, Sasaki DT, Murray BW, O’Leary EC, Sakata ST, Xu W, et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci U S A. 2001;98: 13681–13686. doi: 10.1073/pnas.251194298 11717429

34. Saadeddin A, Babaei-Jadidi R, Spencer-Dene B, Nateri AS. The links between transcription, beta-catenin/JNK signaling, and carcinogenesis. Mol Cancer Res. 2009;7: 1189–1196. doi: 10.1158/1541-7786.MCR-09-0027 19671687

35. Stein B, Angel P, van Dam H, Ponta H, Herrlich P, van der Eb A, et al. Ultraviolet-radiation induced c-jun gene transcription: two AP-1 like binding sites mediate the response. Photochem Photobiol. 1992;55: 409–415. 1561239

36. Nateri AS, Spencer-Dene B, Behrens A. Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature. 2005;437: 281–285. doi: 10.1038/nature03914 16007074

37. Audard V, Grimber G, Elie C, Radenen B, Audebourg A, Letourneur F, et al. Cholestasis is a marker for hepatocellular carcinomas displaying beta-catenin mutations. J Pathol. 2007;212: 345–352. doi: 10.1002/path.2169 17487939

38. Bioulac-Sage P, Cubel G, Balabaud C, Zucman-Rossi J. Revisiting the pathology of resected benign hepatocellular nodules using new immunohistochemical markers. Semin Liver Dis. 2011;31: 91–103. doi: 10.1055/s-0031-1272837 21344354

39. Burchard J, Zhang C, Liu AM, Poon RTP, Lee NPY, Wong K-F, et al. microRNA-122 as a regulator of mitochondrial metabolic gene network in hepatocellular carcinoma. Mol Syst Biol. 2010;6: 402. doi: 10.1038/msb.2010.58 20739924

40. Cagatay T, Ozturk M. P53 mutation as a source of aberrant beta-catenin accumulation in cancer cells. Oncogene. 2002;21: 7971–7980. doi: 10.1038/sj.onc.1205919 12439747

41. Sangkhathat S, Kusafuka T, Miao J, Yoneda A, Nara K, Yamamoto S, et al. In vitro RNA interference against beta-catenin inhibits the proliferation of pediatric hepatic tumors. Int J Oncol. 2006;28: 715–722. 16465377

42. Tward AD, Jones KD, Yant S, Cheung ST, Fan ST, Chen X, et al. Distinct pathways of genomic progression to benign and malignant tumors of the liver. Proc Natl Acad Sci U S A. 2007;104: 14771–14776. doi: 10.1073/pnas.0706578104 17785413

43. Gulbins E, Palmada M, Reichel M, Lüth A, Böhmer C, Amato D, et al. Acid sphingomyelinase-ceramide system mediates effects of antidepressant drugs. Nat Med. 2013;19: 934–938. doi: 10.1038/nm.3214 23770692

44. Gourley SL, Wu FJ, Kiraly DD, Ploski JE, Kedves AT, Duman RS, et al. Regionally specific regulation of ERK MAP kinase in a model of antidepressant-sensitive chronic depression. Biol Psychiatry. 2008;63: 353–359. doi: 10.1016/j.biopsych.2007.07.016 17889834

45. Seki E, Brenner DA, Karin M. A liver full of JNK: signaling in regulation of cell function and disease pathogenesis, and clinical approaches. Gastroenterology. 2012;143: 307–320. doi: 10.1053/j.gastro.2012.06.004 22705006

46. Hagiwara S, Kudo M, Nagai T, Inoue T, Ueshima K, Nishida N, et al. Activation of JNK and high expression level of CD133 predict a poor response to sorafenib in hepatocellular carcinoma. Br J Cancer. 2012;106: 1997–2003. doi: 10.1038/bjc.2012.145 22596232

47. Nagata H, Hatano E, Tada M, Murata M, Kitamura K, Asechi H, et al. Inhibition of c-Jun NH2-terminal kinase switches Smad3 signaling from oncogenesis to tumor- suppression in rat hepatocellular carcinoma. Hepatol. 2009;49: 1944–1953. doi: 10.1002/hep.22860

48. Lesurtel M, Graf R, Aleil B, Walther DJ, Tian Y, Jochum W, et al. Platelet-derived serotonin mediates liver regeneration. Science. 2006;312: 104–107. doi: 10.1126/science.1123842 16601191

49. Gillman PK. Tricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br J Pharmacol. 2007;151: 737–748. doi: 10.1038/sj.bjp.0707253 17471183

50. Diaz SL, Doly S, Narboux-Nême N, Fernández S, Mazot P, Banas SM, et al. 5-HT(2B) receptors are required for serotonin-selective antidepressant actions. Mol Psychiatry. 2012;17: 154–163. doi: 10.1038/mp.2011.159 22158014

51. Jahchan NS, Dudley JT, Mazur PK, Flores N, Yang D, Palmerton A, et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov. 2013;3: 1364–1377. doi: 10.1158/2159-8290.CD-13-0183 24078773

52. Ruddell RG, Oakley F, Hussain Z, Yeung I, Bryan-Lluka LJ, Ramm GA, et al. A role for serotonin (5-HT) in hepatic stellate cell function and liver fibrosis. Am J Pathol. 2006;169: 861–876. doi: 10.2353/ajpath.2006.050767 16936262

53. Mun A-R, Lee S-J, Kim G-B, Kang H-S, Kim J-S, Kim S-J. Fluoxetine-induced apoptosis in hepatocellular carcinoma cells. Anticancer Res. 2013;33: 3691–3697. 24023297

54. Mao X, Hou T, Cao B, Wang W, Li Z, Chen S, et al. The tricyclic antidepressant amitriptyline inhibits D-cyclin transactivation and induces myeloma cell apoptosis by inhibiting histone deacetylases: in vitro and in silico evidence. Mol Pharmacol. 2011;79: 672–680. doi: 10.1124/mol.110.068122 21220410

55. Parker KA, Glaysher S, Hurren J, Knight LA, McCormick D, Suovouri A, et al. The effect of tricyclic antidepressants on cutaneous melanoma cell lines and primary cell cultures. Anticancer Drugs. 2012;23: 65–69. doi: 10.1097/CAD.0b013e32834b1894 21897201

56. Levkovitz Y, Gil-Ad I, Zeldich E, Dayag M, Weizman A. Differential induction of apoptosis by antidepressants in glioma and neuroblastoma cell lines: evidence for p-c-Jun, cytochrome c, and caspase-3 involvement. J Mol Neurosci. 2005;27: 29–42. doi: 10.1385/JMN:27:1:029 16055945

57. Cordero MD, Sánchez-Alcázar JA, Bautista-Ferrufino MR, Carmona-López MI, Illanes M, Ríos MJ, et al. Acute oxidant damage promoted on cancer cells by amitriptyline in comparison with some common chemotherapeutic drugs. Anticancer Drugs. 2010;21: 932–944. doi: 10.1097/CAD.0b013e32833ed5f7 20847644

58. Huang S-MA, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 2009;461: 614–620. doi: 10.1038/nature08356 19759537

59. Ewan K, Pajak B, Stubbs M, Todd H, Barbeau O, Quevedo C, et al. A useful approach to identify novel small-molecule inhibitors of Wnt-dependent transcription. Cancer Res. 2010;70: 5963–5973. doi: 10.1158/0008-5472.CAN-10-1028 20610623

60. Cheung ID, Bagnat M, Ma TP, Datta A, Evason K, Moore JC, et al. Regulation of intrahepatic biliary duct morphogenesis by Claudin 15-like b. Dev Biol. 2012;361: 68–78. doi: 10.1016/j.ydbio.2011.10.004 22020048

61. Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. Stages of embryonic development of the zebrafish. Dev Dyn. 1995;203: 253–310. doi: 10.1002/aja.1002030302 8589427

62. Thermes V, Grabher C, Ristoratore F, Bourrat F, Choulika A, Wittbrodt J, et al. I-SceI meganuclease mediates highly efficient transgenesis in fish. Mech Dev. 2002;118: 91–98. 12351173

63. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinforma Oxf Engl. 2013;29: 15–21. doi: 10.1093/bioinformatics/bts635

64. Lawrence M, Huber W, Pagès H, Aboyoun P, Carlson M, Gentleman R, et al. Software for computing and annotating genomic ranges. PLoS Comput Biol. 2013;9: e1003118. doi: 10.1371/journal.pcbi.1003118 23950696

65. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15: 550. doi: 10.1186/s13059-014-0550-8 25516281

66. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9: 559. doi: 10.1186/1471-2105-9-559 19114008

67. Miller JA, Cai C, Langfelder P, Geschwind DH, Kurian SM, Salomon DR, et al. Strategies for aggregating gene expression data: the collapseRows R function. BMC Bioinformatics. 2011;12: 322. doi: 10.1186/1471-2105-12-322 21816037

68. Langfelder P, Horvath S. Fast R Functions for Robust Correlations and Hierarchical Clustering. J Stat Softw. 2012;46.

69. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; doi: 10.1093/nar/gkv007

70. Durinck S, Spellman PT, Birney E, Huber W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat Protoc. 2009;4: 1184–1191. doi: 10.1038/nprot.2009.97 19617889

71. Durinck S, Moreau Y, Kasprzyk A, Davis S, De Moor B, Brazma A, et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinforma Oxf Engl. 2005;21: 3439–3440. doi: 10.1093/bioinformatics/bti525

72. Husson F, Josse J, Le S, Mazet J. FactoMineR: Multivariate Exploratory Data Analysis and Data Mining [Internet]. 2015. Available: http://factominer.free.fr

73. Maechler M, Rousseeuw P, Struyf A, Hubert M, Hornik K. cluster: Cluster Analysis Basics and Extensions [Internet]. 2015. Available: http://cran.r-project.org/web/packages/cluster/citation.html

74. Warnes GR, Bolker B, Bonebakker L, Gentleman R, Huber W, Liaw A, et al. gplots: Various R Programming Tools for Plotting Data [Internet]. 2015. Available: http://cran.r-project.org/web/packages/gplots/index.html

75. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25: 25–29. doi: 10.1038/75556 10802651

76. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28: 27–30. 10592173

77. Croft D, Mundo AF, Haw R, Milacic M, Weiser J, Wu G, et al. The Reactome pathway knowledgebase. Nucleic Acids Res. 2014;42: D472–477. doi: 10.1093/nar/gkt1102 24243840

78. Field HA, Ober EA, Roeser T, Stainier DYR. Formation of the digestive system in zebrafish. I. Liver morphogenesis. Dev Biol. 2003;253: 279–290. 12645931

79. Yin C, Evason KJ, Maher JJ, Stainier DYR. The basic helix-loop-helix transcription factor, heart and neural crest derivatives expressed transcript 2, marks hepatic stellate cells in zebrafish: analysis of stellate cell entry into the developing liver. Hepatol. 2012;56: 1958–1970. doi: 10.1002/hep.25757

80. Larionov A, Krause A, Miller W. A standard curve based method for relative real time PCR data processing. BMC Bioinformatics. 2005;6: 62. doi: 10.1186/1471-2105-6-62 15780134

81. Evason KJ, Grenert JP, Ferrell LD, Kakar S. Atypical hepatocellular adenoma-like neoplasms with β-catenin activation show cytogenetic alterations similar to well-differentiated hepatocellular carcinomas. Hum Pathol. 2013;44: 750–758. doi: 10.1016/j.humpath.2012.07.019 23084586

82. Horiuchi D, Kusdra L, Huskey NE, Chandriani S, Lenburg ME, Gonzalez-Angulo AM, et al. MYC pathway activation in triple-negative breast cancer is synthetic lethal with CDK inhibition. J Exp Med. 2012;209: 679–696. doi: 10.1084/jem.20111512 22430491

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 7
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#