Abstract

Melanoma is a devastating skin cancer characterized by distinct biological subtypes. Besides frequent mutations in growth- and survival-promoting genes like BRAF and NRAS, melanomas additionally harbor complex non-random genomic alterations. Using an integrative approach, we have analysed genomic and gene expression changes in human melanoma cell lines (N=32) derived from primary tumors and various metastatic sites and investigated the relation to local growth aggressiveness as xenografts in immuno-compromised mice (N=22). Although the vast majority >90% of melanoma models harbored mutations in either BRAF or NRAS, significant differences in subcutaneous growth aggressiveness became obvious. Unsupervised clustering revealed that genomic alterations rather than gene expression data reflected this aggressive phenotype, while no association with histology, stage or metastatic site of the original melanoma was found. Genomic clustering allowed separation of melanoma models into two subgroups with differing local growth aggressiveness in vivo. Regarding genes expressed at significantly altered levels between these subgroups, a surprising correlation with the respective gene doses (>85% accordance) was found. Genes deregulated at the DNA and mRNA level included well-known cancer genes partly already linked to melanoma (RAS genes, PTEN, AURKA, MAPK inhibitors Sprouty/Spred), but also novel candidates like SIPA1 (a Rap1GAP). Pathway mining further supported deregulation of Rap1 signaling in the aggressive subgroup e.g. by additional repression of two Rap1GEFs. Accordingly, siRNA-mediated down-regulation of SIPA1 exerted significant effects on clonogenicity, adherence and migration in aggressive melanoma models. Together our data suggest that an aneuploidy-driven gene expression deregulation drives local aggressiveness in human melanoma.

Highlights

  • Melanomas account for 4 to 5% of all cancers and represent currently the 6th leading cancer type in the USA [1]

  • Melanoma cells constitutively harbor - besides welldefined mutations in certain proto-oncogenes like BRAF and NRAS - non-random genomic alterations reflecting chromosomal instability (CIN) [11, 12]

  • Using in vivo growth aggressiveness as a grouping criterion, we provide strong evidence that aneuploidy-mediated gene expression alterations are key drivers of aggressive melanoma growth

Read more

Summary

Introduction

Melanomas account for 4 to 5% of all cancers and represent currently the 6th leading cancer type in the USA [1]. Recent discoveries have markedly improved our understanding of the molecular changes underlying malignant progression of melanomas including mainly alterations in proliferation, survival and cell death signaling pathways [3,4,5]. The RAF/MEK/ERK and PI3K/AKT are two major signaling pathways constitutively activated in up to 90% of melanomas. The AKT/mTOR pathway might be activated mainly by loss-of-function mutations or deletions of the inhibiting phosphatase PTEN. Typical impairment of senescence due to mutations, deletions or methylation of p16INK4/CDKN2A occurs in 30 to 70% of melanomas. Oncogenic proteins in melanoma include e.g. members of the bcl-2 protein family, cyclin D1, and several transcription factors like the lineage-specific oncogene MITF (for detailed reviews on these molecular changes see [6,7,8])

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call