Specific c-Jun target genes in malignant melanoma

ABSTRACT A fundamental event in the development and progression of malignant melanoma is the de-regulation of cancer-relevant transcription factors. We recently showed that c-Jun is a main regulator of melanoma progression and, thus, is the most important member of the AP-1 transcription factor family in this disease. Surprisingly, no cancer-related specific c-Jun target genes in melanoma were described in the literature, so far. Therefore, we focused on pre-existing ChIP-Seq data (Encyclopedia of DNA Elements) of 3 different non-melanoma cell lines to screen direct c-Jun target genes. Here, a specific c-Jun antibody to immunoprecipitate the associated promoter DNA was used. Consequently, we identified 44 direct c-Jun targets and a detailed analysis of 6 selected genes confirmed their deregulation in malignant melanoma. The identified genes were differentially regulated comparing 4 melanoma cell lines and normal human melanocytes and we confirmed their c-Jun dependency. Direct interaction between c-Jun and the promoter/enhancer regions of the identified genes was confirmed by us via ChIP experiments. Interestingly, we revealed that the direct regulation of target gene expression via c-Jun can be independent of the existence of the classical AP-1 (5´-TGA(C/G)TCA-3´) consensus sequence allowing for the subsequent down- or up-regulation of the expression of these cancer-relevant genes. In summary, the results of this study indicate that c-Jun plays a crucial role in the development and progression of malignant melanoma via direct regulation of cancer-relevant target genes and that inhibition of direct c-Jun targets through inhibition of c-Jun is a potential novel therapeutic option for treatment of malignant melanoma.

[1]  H. Simon,et al.  Autophagy suppresses melanoma tumorigenesis by inducing senescence , 2014, Autophagy.

[2]  Charles Elkan,et al.  Fitting a Mixture Model By Expectation Maximization To Discover Motifs In Biopolymer , 1994, ISMB.

[3]  T. Kerppola,et al.  Close encounters of many kinds: Fos-Jun interactions that mediate transcription regulatory specificity , 2001, Oncogene.

[4]  J. McCubrey,et al.  Nectin like-5 overexpression correlates with the malignant phenotype in cutaneous melanoma , 2012, Oncotarget.

[5]  M. Karin,et al.  AP-1 in cell proliferation and survival , 2001, Oncogene.

[6]  E. Wagner,et al.  Control of cell cycle progression by c-Jun is p53 dependent. , 1999, Genes & development.

[7]  Richard A Young,et al.  Chromatin immunoprecipitation and microarray-based analysis of protein location , 2006, Nature Protocols.

[8]  J. Barrett,et al.  Pathway-Based Analysis of a Melanoma Genome-Wide Association Study: Analysis of Genes Related to Tumour-Immunosuppression , 2011, PloS one.

[9]  M. Bar‐eli,et al.  Expression profiling of Galectin-3-depleted melanoma cells reveals its major role in melanoma cell plasticity and vasculogenic mimicry. , 2008, The American journal of pathology.

[10]  Juan Wang,et al.  Aberrant Expression of Beclin-1 and LC3 Correlates with Poor Prognosis of Human Hypopharyngeal Squamous Cell Carcinoma , 2013, PloS one.

[11]  O. Wolkenhauer,et al.  Regulation of cell cycle checkpoint kinase WEE1 by miR-195 in malignant melanoma , 2013, Oncogene.

[12]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[13]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[14]  E. Wagner,et al.  JunB suppresses cell proliferation by transcriptional activation of p16INK4a expression , 2000, The EMBO journal.

[15]  K. Milde-Langosch,et al.  Expression pattern of the AP‐1 family in breast cancer: Association of fosB expression with a well‐differentiated, receptor‐positive tumor phenotype , 1999, International journal of cancer.

[16]  Dong-hao Wu,et al.  Autophagic LC3B overexpression correlates with malignant progression and predicts a poor prognosis in hepatocellular carcinoma , 2014, Tumor Biology.

[17]  W. Nishio,et al.  The role of Necl-5 in the invasive activity of lung adenocarcinoma. , 2013, Experimental and molecular pathology.

[18]  Paola Baldassari,et al.  NFATc2 is a potential therapeutic target in human melanoma. , 2012, The Journal of investigative dermatology.

[19]  F. Meyskens,et al.  During human melanoma progression AP-1 binding pairs are altered with loss of c-Jun in vitro. , 2004, Pigment cell research.

[20]  David Bryant,et al.  DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists , 2007, Nucleic Acids Res..

[21]  Tao Lu,et al.  Role of ChIP-seq in the discovery of transcription factor binding sites, differential gene regulation mechanism, epigenetic marks and beyond , 2014, Cell cycle.

[22]  E. Bossy‐Wetzel,et al.  Cell cycle‐dependent variations in c‐Jun and JunB phosphorylation: a role in the control of cyclin D1 expression , 2000, The EMBO journal.

[23]  M. Karin,et al.  Transcriptional interference between c-Jun and the glucocorticoid receptor: Mutual inhibition of DNA binding due to direct protein-protein interaction , 1990, Cell.

[24]  E. Shaulian AP-1--The Jun proteins: Oncogenes or tumor suppressors in disguise? , 2010, Cellular signalling.

[25]  A. Bosserhoff,et al.  Death inducer-obliterator 1 (Dido1) is a BMP target gene and promotes BMP-induced melanoma progression , 2013, Oncogene.

[26]  Brad T. Sherman,et al.  The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists , 2007, Genome Biology.

[27]  A. Bosserhoff,et al.  AP-1/c-Jun transcription factors: regulation and function in malignant melanoma. , 2014, European journal of cell biology.

[28]  A. Bosserhoff,et al.  MicroRNA miR-125b controls melanoma progression by direct regulation of c-Jun protein expression , 2013, Oncogene.

[29]  C. Weiss,et al.  Deregulated Repression of c-Jun Provides a Potential Link to its Role in Tumorigenesis , 2004, Cell cycle.

[30]  D. Louis,et al.  CD155/PVR plays a key role in cell motility during tumor cell invasion and migration , 2004, BMC Cancer.

[31]  A. Bosserhoff,et al.  Functional role of MIA in melanocytes and early development of melanoma , 2004, Oncogene.

[32]  S. Grässel,et al.  The transcription factor AP-2ɛ regulates CXCL1 during cartilage development and in osteoarthritis. , 2011, Osteoarthritis and cartilage.

[33]  Michael D. Taylor,et al.  Integrated genomic analysis identifies the mitotic checkpoint kinase WEE1 as a novel therapeutic target in medulloblastoma , 2014, Molecular Cancer.

[34]  E. Wagner,et al.  AP-1: a double-edged sword in tumorigenesis , 2003, Nature Reviews Cancer.

[35]  A. Bosserhoff,et al.  Post‐transcriptional regulation controlled by E‐cadherin is important for c‐Jun activity in melanoma , 2011, Pigment cell & melanoma research.

[36]  S. Choi,et al.  RGS16 and FosB underexpressed in pancreatic cancer with lymph node metastasis promote tumor progression , 2010, Tumor Biology.

[37]  A. Bosserhoff,et al.  ETS‐1/RhoC signaling regulates the transcription factor c‐Jun in melanoma , 2012, International journal of cancer.

[38]  V. Sondak,et al.  Inhibition of Wee1, AKT, and CDK4 Underlies the Efficacy of the HSP90 Inhibitor XL888 in an In Vivo Model of NRAS-Mutant Melanoma , 2013, Molecular Cancer Therapeutics.

[39]  P. Vogt,et al.  Jun, the oncoprotein , 2001, Oncogene.

[40]  A. Ghaderi,et al.  Tumour suppressive effects of WEE1 gene silencing in breast cancer cells. , 2013, Asian Pacific journal of cancer prevention : APJCP.

[41]  D. McConkey,et al.  Galectin-3 contributes to melanoma growth and metastasis via regulation of NFAT1 and autotaxin. , 2012, Cancer research.

[42]  Z. Krawczyk,et al.  Melanoma-associated genes, MXI1, FN1, and NME1, are hypoxia responsive in murine and human melanoma cells , 2011, Melanoma research.

[43]  V. A. Flørenes,et al.  Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. , 2014, Gynecologic oncology.

[44]  E. Wagner,et al.  AP-1 in mouse development and tumorigenesis , 2001, Oncogene.