#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Cross-Species Array Comparative Genomic Hybridization Identifies Novel Oncogenic Events in Zebrafish and Human Embryonal Rhabdomyosarcoma


Human cancer genomes are highly complex, making it challenging to identify specific drivers of cancer growth, progression, and tumor maintenance. To bypass this obstacle, we have applied array comparative genomic hybridization (array CGH) to zebrafish embryonal rhabdomyosaroma (ERMS) and utilized cross-species comparison to rapidly identify genomic copy number aberrations and novel candidate oncogenes in human disease. Zebrafish ERMS contain small, focal regions of low-copy amplification. These same regions were commonly amplified in human disease. For example, 16 of 19 chromosomal gains identified in zebrafish ERMS also exhibited focal, low-copy gains in human disease. Genes found in amplified genomic regions were assessed for functional roles in promoting continued tumor growth in human and zebrafish ERMS – identifying critical genes associated with tumor maintenance. Knockdown studies identified important roles for Cyclin D2 (CCND2), Homeobox Protein C6 (HOXC6) and PlexinA1 (PLXNA1) in human ERMS cell proliferation. PLXNA1 knockdown also enhanced differentiation, reduced migration, and altered anchorage-independent growth. By contrast, chemical inhibition of vascular endothelial growth factor (VEGF) signaling reduced angiogenesis and tumor size in ERMS-bearing zebrafish. Importantly, VEGFA expression correlated with poor clinical outcome in patients with ERMS, implicating inhibitors of the VEGF pathway as a promising therapy for improving patient survival. Our results demonstrate the utility of array CGH and cross-species comparisons to identify candidate oncogenes essential for the pathogenesis of human cancer.


Vyšlo v časopise: Cross-Species Array Comparative Genomic Hybridization Identifies Novel Oncogenic Events in Zebrafish and Human Embryonal Rhabdomyosarcoma. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003727
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003727

Souhrn

Human cancer genomes are highly complex, making it challenging to identify specific drivers of cancer growth, progression, and tumor maintenance. To bypass this obstacle, we have applied array comparative genomic hybridization (array CGH) to zebrafish embryonal rhabdomyosaroma (ERMS) and utilized cross-species comparison to rapidly identify genomic copy number aberrations and novel candidate oncogenes in human disease. Zebrafish ERMS contain small, focal regions of low-copy amplification. These same regions were commonly amplified in human disease. For example, 16 of 19 chromosomal gains identified in zebrafish ERMS also exhibited focal, low-copy gains in human disease. Genes found in amplified genomic regions were assessed for functional roles in promoting continued tumor growth in human and zebrafish ERMS – identifying critical genes associated with tumor maintenance. Knockdown studies identified important roles for Cyclin D2 (CCND2), Homeobox Protein C6 (HOXC6) and PlexinA1 (PLXNA1) in human ERMS cell proliferation. PLXNA1 knockdown also enhanced differentiation, reduced migration, and altered anchorage-independent growth. By contrast, chemical inhibition of vascular endothelial growth factor (VEGF) signaling reduced angiogenesis and tumor size in ERMS-bearing zebrafish. Importantly, VEGFA expression correlated with poor clinical outcome in patients with ERMS, implicating inhibitors of the VEGF pathway as a promising therapy for improving patient survival. Our results demonstrate the utility of array CGH and cross-species comparisons to identify candidate oncogenes essential for the pathogenesis of human cancer.


Zdroje

1. LinaberyAM, RossJA (2008) Trends in childhood cancer incidence in the U.S. (1992–2004). Cancer 112: 416–432.

2. ChenY, TakitaJ, HiwatariM, IgarashiT, HanadaR, et al. (2006) Mutations of the PTPN11 and RAS genes in rhabdomyosarcoma and pediatric hematological malignancies. Genes Chromosomes Cancer 45: 583–591.

3. HettmerS, LiuJ, MillerCM, LindsayMC, SparksCA, et al. (2011) Sarcomas induced in discrete subsets of prospectively isolated skeletal muscle cells. Proc Natl Acad Sci U S A 108: 20002–20007.

4. LangenauDM, KeefeMD, StorerNY, GuyonJR, KutokJL, et al. (2007) Effects of RAS on the genesis of embryonal rhabdomyosarcoma. Genes Dev 21: 1382–1395.

5. PaulsonV, ChandlerG, RakhejaD, GalindoRL, WilsonK, et al. (2011) High-resolution array CGH identifies common mechanisms that drive embryonal rhabdomyosarcoma pathogenesis. Genes Chromosomes Cancer 50: 397–408.

6. BridgeJA, LiuJ, QualmanSJ, SuijkerbuijkR, WengerG, et al. (2002) Genomic gains and losses are similar in genetic and histologic subsets of rhabdomyosarcoma, whereas amplification predominates in embryonal with anaplasia and alveolar subtypes. Genes Chromosomes Cancer 33: 310–321.

7. GoldsteinM, MellerI, IssakovJ, Orr-UrtregerA (2006) Novel genes implicated in embryonal, alveolar, and pleomorphic rhabdomyosarcoma: a cytogenetic and molecular analysis of primary tumors. Neoplasia 8: 332–343.

8. MissiagliaE, SelfeJ, HamdiM, WilliamsonD, SchaafG, et al. (2009) Genomic imbalances in rhabdomyosarcoma cell lines affect expression of genes frequently altered in primary tumors: an approach to identify candidate genes involved in tumor development. Genes Chromosomes Cancer 48: 455–467.

9. PanditaA, ZielenskaM, ThornerP, BayaniJ, GodboutR, et al. (1999) Application of comparative genomic hybridization, spectral karyotyping, and microarray analysis in the identification of subtype-specific patterns of genomic changes in rhabdomyosarcoma. Neoplasia 1: 262–275.

10. DavariP, HebertJL, AlbertsonDG, HueyB, RoyR, et al. (2010) Loss of Blm enhances basal cell carcinoma and rhabdomyosarcoma tumorigenesis in Ptch1+/− mice. Carcinogenesis 31: 968–973.

11. RubinBP, NishijoK, ChenHI, YiX, SchuetzeDP, et al. (2011) Evidence for an unanticipated relationship between undifferentiated pleomorphic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell 19: 177–191.

12. GoesslingW, NorthTE, LoewerS, LordAM, LeeS, et al. (2009) Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136: 1136–1147.

13. LamSH, WuYL, VegaVB, MillerLD, SpitsbergenJ, et al. (2006) Conservation of gene expression signatures between zebrafish and human liver tumors and tumor progression. Nat Biotechnol 24: 73–75.

14. LiuS, LeachSD (2011) Zebrafish models for cancer. Annu Rev Pathol 6: 71–93.

15. PattonEE, WidlundHR, KutokJL, KopaniKR, AmatrudaJF, et al. (2005) BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma. Curr Biol 15: 249–254.

16. WhiteRM, CechJ, RatanasirintrawootS, LinCY, RahlPB, et al. (2011) DHODH modulates transcriptional elongation in the neural crest and melanoma. Nature 471: 518–522.

17. StrattonMR, FisherC, GustersonBA, CooperCS (1989) Detection of point mutations in N-ras and K-ras genes of human embryonal rhabdomyosarcomas using oligonucleotide probes and the polymerase chain reaction. Cancer Res 49: 6324–6327.

18. FreemanJL, CeolC, FengH, LangenauDM, BelairC, et al. (2009) Construction and application of a zebrafish array comparative genomic hybridization platform. Genes Chromosomes Cancer 48: 155–170.

19. ZhangG, HoerschS, AmsterdamA, WhittakerCA, LeesJA, et al. (2010) Highly aneuploid zebrafish malignant peripheral nerve sheath tumors have genetic alterations similar to human cancers. Proc Natl Acad Sci U S A 107: 16940–16945.

20. RudnerLA, BrownKH, DobrinskiKP, BradleyDF, GarciaMI, et al. (2011) Shared acquired genomic changes in zebrafish and human T-ALL. Oncogene 30: 4289–4296.

21. IgnatiusMS, ChenE, ElpekNM, FullerAZ, TenenteIM, et al. (2012) In vivo imaging of tumor-propagating cells, regional tumor heterogeneity, and dynamic cell movements in embryonal rhabdomyosarcoma. Cancer Cell 21: 680–693.

22. LeX, LangenauDM, KeefeMD, KutokJL, NeubergDS, et al. (2007) Heat shock-inducible Cre/Lox approaches to induce diverse types of tumors and hyperplasia in transgenic zebrafish. Proc Natl Acad Sci U S A 104: 9410–9415.

23. DavicioniE, AndersonJR, BuckleyJD, MeyerWH, TricheTJ (2010) Gene expression profiling for survival prediction in pediatric rhabdomyosarcomas: a report from the children's oncology group. J Clin Oncol 28: 1240–1246.

24. MerlinoG, HelmanLJ (1999) Rhabdomyosarcoma–working out the pathways. Oncogene 18: 5340–5348.

25. TapscottSJ, ThayerMJ, WeintraubH (1993) Deficiency in rhabdomyosarcomas of a factor required for MyoD activity and myogenesis. Science 259: 1450–1453.

26. LawsonND, WeinsteinBM (2002) In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol 248: 307–318.

27. RaneyRB, AndersonJR, BarrFG, DonaldsonSS, PappoAS, et al. (2001) Rhabdomyosarcoma and undifferentiated sarcoma in the first two decades of life: a selective review of intergroup rhabdomyosarcoma study group experience and rationale for Intergroup Rhabdomyosarcoma Study V. J Pediatr Hematol Oncol 23: 215–220.

28. YangXR, NgD, AlcortaDA, LiebschNJ, SheridanE, et al. (2009) T (brachyury) gene duplication confers major susceptibility to familial chordoma. Nat Genet 41: 1176–1178.

29. LahortigaI, De KeersmaeckerK, Van VlierbergheP, GrauxC, CauwelierB, et al. (2007) Duplication of the MYB oncogene in T cell acute lymphoblastic leukemia. Nat Genet 39: 593–595.

30. NgCK, CookeSL, HoweK, NewmanS, XianJ, et al. (2011) The role of tandem duplicator phenotype in tumour evolution in high-grade serous ovarian cancer. J Pathol 226: 703–12.

31. McCabeCD, SpyropoulosDD, MartinD, MorenoCS (2008) Genome-wide analysis of the homeobox C6 transcriptional network in prostate cancer. Cancer Res 68: 1988–1996.

32. MusgroveEA, CaldonCE, BarracloughJ, StoneA, SutherlandRL (2011) Cyclin D as a therapeutic target in cancer. Nat Rev Cancer 11: 558–572.

33. BuschgesR, WeberRG, ActorB, LichterP, CollinsVP, et al. (1999) Amplification and expression of cyclin D genes (CCND1, CCND2 and CCND3) in human malignant gliomas. Brain Pathol 9: 435–442; discussion 432–433.

34. KrugerRP, AurandtJ, GuanKL (2005) Semaphorins command cells to move. Nat Rev Mol Cell Biol 6: 789–800.

35. NegishiM, OinumaI, KatohH (2005) Plexins: axon guidance and signal transduction. Cell Mol Life Sci 62: 1363–1371.

36. GranzieroL, CircostaP, ScielzoC, FrisaldiE, StellaS, et al. (2003) CD100/Plexin-B1 interactions sustain proliferation and survival of normal and leukemic CD5+ B lymphocytes. Blood 101: 1962–1969.

37. ValenteG, NicotraG, ArrondiniM, CastinoR, CapparucciaL, et al. (2009) Co-expression of plexin-B1 and Met in human breast and ovary tumours enhances the risk of progression. Cell Oncol 31: 423–436.

38. YeS, HaoX, ZhouT, WuM, WeiJ, et al. (2010) Plexin-B1 silencing inhibits ovarian cancer cell migration and invasion. BMC Cancer 10: 611.

39. CatalanoA, LazzariniR, Di NuzzoS, OrciariS, ProcopioA (2009) The plexin-A1 receptor activates vascular endothelial growth factor-receptor 2 and nuclear factor-kappaB to mediate survival and anchorage-independent growth of malignant mesothelioma cells. Cancer Res 69: 1485–1493.

40. KasebAO, MorrisJS, HassanMM, SiddiquiAM, LinE, et al. (2011) Clinical and prognostic implications of plasma insulin-like growth factor-1 and vascular endothelial growth factor in patients with hepatocellular carcinoma. J Clin Oncol 29: 3892–3899.

41. MaaeE, OlsenDA, SteffensenKD, JakobsenEH, BrandslundI, et al. (2012) Prognostic impact of placenta growth factor and vascular endothelial growth factor A in patients with breast cancer. Breast Cancer Res Treat 133: 257–65.

42. PrinsMJ, VerhageRJ, Ten KateFJ, van HillegersbergR (2012) Cyclooxygenase Isoenzyme-2 and Vascular Endothelial Growth Factor are Associated with Poor Prognosis in Esophageal Adenocarcinoma. J Gastrointest Surg 16: 956–66.

43. MarisJM, CourtrightJ, HoughtonPJ, MortonCL, GorlickR, et al. (2008) Initial testing of the VEGFR inhibitor AZD2171 by the pediatric preclinical testing program. Pediatr Blood Cancer 50: 581–587.

44. BoucheO, Maindrault-GoebelF, DucreuxM, LledoG, AndreT, et al. (2011) Phase II trial of weekly alternating sequential BIBF 1120 and afatinib for advanced colorectal cancer. Anticancer Res 31: 2271–2281.

45. FountzilasG, FragkoulidiA, Kalogera-FountzilaA, NikolaidouM, BobosM, et al. (2010) A phase II study of sunitinib in patients with recurrent and/or metastatic non-nasopharyngeal head and neck cancer. Cancer Chemother Pharmacol 65: 649–660.

46. IwamotoFM, LambornKR, RobinsHI, MehtaMP, ChangSM, et al. (2010) Phase II trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06-02). Neuro Oncol 12: 855–861.

47. SmithAC, RaimondiAR, SalthouseCD, IgnatiusMS, BlackburnJS, et al. (2010) High-throughput cell transplantation establishes that tumor-initiating cells are abundant in zebrafish T-cell acute lymphoblastic leukemia. Blood 115: 3296–3303.

48. GuyonJR, MosleyAN, ZhouY, O'BrienKF, ShengX, et al. (2003) The dystrophin associated protein complex in zebrafish. Hum Mol Genet 12: 601–615.

49. WeidnerN, SempleJP, WelchWR, FolkmanJ (1991) Tumor angiogenesis and metastasis–correlation in invasive breast carcinoma. N Engl J Med 324: 1–8.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 8
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#