Horizontal propagation of EMT, stemness, and metastasis by transferring the MAP17 protein through secreted vesicles

José Manuel García-Heredia, Daniel Otero-Albiol, Marco Pérez, Elena Pérez-Castejón, Sandra Muñoz-Galván and Amancio Carnero; Instituto de Biomedicina de Sevilla (IBIS/HUVR/Universidad de Sevilla/CSIC); CIBERONC
Published in Cancer
Like

MAP17 (PDZK1IP1, DD96, SPAP) [1-3] is a small (approximately 17 kDa), nonglycosylated protein usually localized to the plasma membrane and associated with areas of cell-cell contact [4]. Although its expression in non-tumoral cells is restricted to specific epithelial cell populations in kidney [4], more than 50% of advanced tumors exhibit significant expression of MAP17 [1, 3, 5, 6]. High MAP17 levels appear in most human carcinomas and in other non-epithelial neoplasias, such as glioblastomas or lymphomas [4, 7]. This protein is also associated with tumor progression, since it regulates cellular transformation and malignancy [1, 8, 9], correlating its expression with an increase in cell de-differentiation [8-10]. It has also been recently shown that the expression of MAP17 is not restricted to cancer, appearing upregulated in chronic inflammatory diseases and induces the transcription of genes related to inflammatory response, such as HLAs, IL-6 and NFAT2 [6, 11].

Although MAP17 has no enzymatic or transcriptional activity, it exerts its described roles through the regulation of several cell signaling pathways. MAP17 overexpression activates NOTCH pathway by sequestrating NUMB, a known inhibitor of NOTCH, allowing a higher activation of the pathway [12, 13].

During with tumor progression, MAP17 levels increase due to its promoter demethylation in cancer cells. The increased MAP17 levels in tumors induce NOTCH pathway activation regulating the expression or transcriptional repression of specific microRNAs producing multiple changes in gene expression. As a consequence,  the epithelial mesenchymal transition, EMT, and stemness are induced, greatly increasing the metastatic potential of these cells both in vitro and in vivo. Additionally, MAP17 overexpression also increased extracellular vesicle secretion, and MAP17 itself was secreted as a cargo in these particles, inducing EMT and stemness phenotypes in the recipient cells. Therefore, MAP17 expression epigenetically enhances the horizontal propagation of EMT and metastatic phenotypes by transferring itself between subsets of neoplastic cells. Finally, elimination of MAP17 or exosomes from the microenvironment abrogates the induction of EMT/stemness transcription.

Altogether, our work provides a totally unknown functional circuit between the horizontal propagation, NOTCH pathway, miRNAs regulation, EMT-CSC pool and metastases and the oncogene MAP17, overexpresed in around a 50% of human breast tumors. This may provide, in the future, a new form to attack breast cancer metastasis. Furthermore, the mechanism might be broader since most tumor types, at advances stages, also present high levels of MAP17 [6,8].

  1. Guijarro, M. V., et al. MAP17 overexpression is a common characteristic of carcinomas. Carcinogenesis 2007; 28: 1646-1652
  2. Kocher, O., et al. Identification of a novel gene, selectively up-regulated in human carcinomas, using the differential display technique. Clinical Cancer Research 1995; 1: 1209-1215
  3. Wang, N., et al. Screening and Identification of Distant Metastasis-Related Differentially Expressed Genes in Human Squamous Cell Lung Carcinoma. The Anatomical Record: Advances in Integrative Anatomy and Evolutionary Biology 2012; 295: 748-757
  4. Kocher, O., et al.Identification and partial characterization of a novel membrane-associated protein (MAP17) up-regulated in human carcinomas and modulating cell replication and tumor growth. The American Journal of Pathology 1996; 149: 493-500
  5. Carnero, A. MAP17, a ROS-dependent oncogene. Frontiers in Oncology 2012; 2: 112
  6. García-Heredia, J. M., Carnero, A. Dr. Jekyll and Mr. Hyde: MAP17’s up-regulation, a crosspoint in cancer and inflammatory diseases. Molecular Cancer 2018; 17: 80
  7. Guijarro, M. V., et al. MAP17 inhibits Myc-induced apoptosis through PI3K/AKT pathway activation. Carcinogenesis 2007; 28: 2443-2450
  8. Carnero, A. MAP17 and the double-edged sword of ROS. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 2012; 1826: 44-52
  9. Perez, M., et al. MAP17 and SGLT1 Protein Expression Levels as Prognostic Markers for Cervical Tumor Patient Survival. PLOS ONE 2013; 8: e56169
  10. Guijarro, M. V., et al. p38[alpha] limits the contribution of MAP17 to cancer progression in breast tumors. Oncogene 2012; 31: 4447-4459
  11. Garcia-Heredia, J. M., Carnero, A. The cargo protein MAP17 (PDZK1IP1) regulates the immune microenvironment. Oncotarget 2017; 8: 98580-98597
  12. Garcia-Heredia, J. M., et al. The cargo protein MAP17 (PDZK1IP1) regulates the cancer stem cell pool activating the Notch pathway by abducting NUMB. Clinical Cancer Research 2017; 23: 3871-3883
  13. Wang, Z., et al. Emerging Role of Notch in Stem Cells and Cancer. Cancer letters 2009; 279: 8-12

Link:  https://www.nature.com/articles/s41389-020-00280-0

Please sign in or register for FREE

If you are a registered user on Research Communities by Springer Nature, please sign in

Subscribe to the Topic

Cancer Biology
Life Sciences > Biological Sciences > Cancer Biology
  • Oncogenesis Oncogenesis

    A peer-reviewed open access online journal that publishes articles exploring mechanistic insight and molecular basis of cancer and related phenomena. It seeks to promote diverse and integrated areas of molecular biology, cell biology, oncology, and genetics.