Central Role of CEMIP in Tumorigenesis and Its Potential as Therapeutic Target

Li Li, Lin-Hai Yan, Shwetha Manoj, Ying Li, Lu Lu, Li Li, Lin-Hai Yan, Shwetha Manoj, Ying Li, Lu Lu

Abstract

CEMIP (KIAA1199) was identified as migratory indicator protein which had been crudely studied in the last decade. Firstly its mutation site was reported to cause hearing loss due to the folding change of protein structure, meanwhile the over-expression of CEMIP referred to dreadful invasion and uncontrolled proliferation of tumor with distant metastasis, dedifferentiation, and limited survival opportunity of patients. Especially, over-expressed CEMIP also protected malignant tumor from strict microenvironment in hypoxia, low glucose and cracked barrier, leading to enhanced adaptability of tumor by stimulating the Wnt, EGFR, FGFR pathway. Here, we intend to elaborate the clinical function and dysregulation of CEMIP under the tumorous circumstance since CEMIP plays an important role in cytokine pathway and its over-expression in tumors provide a novel target for individual therapy. Targeting CEMIP would thereby dysregulate the cytokine pathway which would in turn, decide the growth and death of the vicious tumour cells.

Keywords: CEMIP/KIAA1199; Cancer; Cytokine Signaling pathway; Hyaluronic acid..

Conflict of interest statement

Competing Interests: None declared.

Figures

Figure 1
Figure 1
The Simple Wnt/β-catenin pathway involving CEMIP. The Wnt signals will be captured by Frizzled receptors, then they are passed to β-catenin after forming complex with DSH. Over-expression of CEMIP will emphasize this formation and stimulate movement of DSH-Wnt complex towards nucleus. Also, the expression of nucleus-settled β-catenin positively induce the expression of CEMIP. Once bond to TCF complex, the transcription of small secreted protein would be started. Besides, CEMIP also stimulated the ITR3 leading the increase of Ca2+, which was also one of the Wnt signaling factor leading to intracellular ions disordered.
Figure 2
Figure 2
The EGFR signaling pathway involving CEMIP mediation. When EGF signals firstly attach HER2 receptor, heterodimerization and phosphorylation of HER2 and HER3 will be automatically processed leading to enablement of EGFR signaling pathway. The signal will head into two branches, descending activation and degradation where over-expressed CEMIP acts as connected junction. The light purple color represents CEMIP and its simple protein structure with specific binding site, wherein survival signal Src targeted 60AA near N-terminal, and NRG1 bond to N-terminal. Src branch towards Semaphorin-3A-meidated survive and NRG1 lead to proliferation through activation of MEK1. Plexin A2 bond to G8 domain of C-terminal initiating suppression of apoptosis. The dashed arrows describe the EGF signaling routine, so do the curved arrows. Thick line from Plexin A2 to Semaphorin 3A means it cuts off the delivery of apoptotic signal to Semaphorin 3A. The participation of over-expressed CEMIP also prevent EGF signals from consumption of lysosomal enzyme (LAMP2).

References

    1. Raish M, Khurshid M, Ansari MA. et al. Analysis of molecular cytogenetic alterations in uterine leiomyosarcoma by array-based comparative genomic hybridization. J Cancer Res Clin Oncol. 2012;138:1173–1186.
    1. Dash DP, Silvestri G, Hughes AE. Fine mapping of the keratoconus with cataract locus on chromosome 15q and candidate gene analysis. Mol Vis. 2006;12:499–505.
    1. Birkenkamp-Demtroder K, Maghnouj A, Mansilla F. et al. Repression of KIAA1199 attenuates wnt-signalling and decreases the proliferation of colon cancer cells. Br J Cancer. 2011;105:552–561.
    1. He QY, Liu XH, Li Q. et al. G8: A novel domain associated with polycystic kidney disease and non-syndromic hearing loss. Bioinformatics. 2006;22:2189–2191.
    1. Yoshida H, Nagaoka A, Nakamura S. et al. N-terminal signal sequence is required for cellular trafficking and hyaluronan-depolymerization of KIAA1199. FEBS Lett. 2014;588:111–116.
    1. Abe S, Usami S, Nakamura Y. Mutations in the gene encoding KIAA1199 protein, an inner-ear protein expressed in deiters' cells and the fibrocytes, as the cause of nonsyndromic hearing loss. J Hum Genet. 2003;48:564–570.
    1. Guo J, Cheng H, Zhao S. et al. Gg: A domain involved in phage LTF apparatus and implicated in human MEB and non-syndromic hearing loss diseases. FEBS Lett. 2006;580:581–584.
    1. Michishita E, Garces G, Barrett JC. et al. Upregulation of the KIAA1199 gene is associated with cellular mortality. Cancer Lett. 2006;239:71–77.
    1. Usami S, Takumi Y, Suzuki N. et al. The localization of proteins encoded by CRYM, KIAA1199, UBA52, COL9A3, and COL9A1, genes highly expressed in the cochlea. Neuroscience. 2008;154:22–28.
    1. Tiwari A, Schneider M, Fiorino A. et al. Early insights into the function of KIAA1199, a markedly overexpressed protein in human colorectal tumors. PloS One. 2013;8:e69473.
    1. Fink SP, Myeroff LL, Kariv R. et al. Induction of KIAA1199/CEMIP is associated with colon cancer phenotype and poor patient survival. Oncotarget. 2015;6:30500–30515.
    1. Kuscu C, Evensen N, Kim D. et al. Transcriptional and epigenetic regulation of KIAA1199 gene expression in human breast cancer. PloS One. 2012;7:e44661.
    1. Evensen NA, Kuscu C, Nguyen HL. et al. Unraveling the role of KIAA1199, a novel endoplasmic reticulum protein, in cancer cell migration. J Natl Cancer Inst. 2013;105:1402–1416.
    1. Yang X, Qiu P, Chen B. et al. KIAA1199 as a potential diagnostic biomarker of rheumatoid arthritis related to angiogenesis. Arthritis Res Ther. 2015;17:140.
    1. van Heumen BW, Roelofs HM, te Morsche RH. et al. Duodenal mucosal risk markers in patients with familial adenomatous polyposis: Effects of celecoxib/ursodeoxycholic acid co-treatment and comparison with patient controls. Orphanet J Rare Dis. 2013;8:181.
    1. Yoshino Y, Ishisaka M, Tsuruma K. et al. Distribution and function of hyaluronan binding protein involved in hyaluronan depolymerization (HYBID, KIAA1199) in the mouse central nervous system. Neuroscience. 2017;347:1–10.
    1. Terashima M, Fujita Y, Togashi Y. et al. KIAA1199 interacts with glycogen phosphorylase kinase beta-subunit (PHKB) to promote glycogen breakdown and cancer cell survival. Oncotarget. 2014;5:7040–7050.
    1. Zhang Y, Jia S, Jiang WG. KIAA1199 and its biological role in human cancer and cancer cells (review) Oncol Rep. 2014;31:1503–1508.
    1. Xu J, Liu Y, Wang X. et al. Association between KIAA1199 overexpression and tumor invasion, TNM stage, and poor prognosis in colorectal cancer. Int J Clin Exp Pathol. 2015;8:2909–2918.
    1. Hosoya M, Fujioka M. Distinct expression pattern of a deafness gene, KIAA1199, in a primate cochlea. Biomed Res Int. 2016;2016:1781894.
    1. Yoshida H, Nagaoka A, Nakamura S. et al. Murine homologue of the human KIAA1199 is implicated in hyaluronan binding and depolymerization. FEBS Open Bio. 2013;3:352–356.
    1. Usami S, Wagatsuma M, Fukuoka H. et al. The responsible genes in Japanese deafness patients and clinical application using invader assay. Acta Otolaryngol. 2008;128:446–454.
    1. Cheng XB, Kohi S, Koga A. et al. Hyaluronan stimulates pancreatic cancer cell motility. Oncotarget. 2016;7:4829–4840.
    1. Yoshida H, Nagaoka A, Kusaka-Kikushima A. et al. KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization. Proc Natl Acad Sci U S A. 2013;110:5612–5617.
    1. Soroosh A, Albeiroti S, West GA. et al. Crohn's disease fibroblasts overproduce the novel protein KIAA1199 to create proinflammatory hyaluronan fragments. Cell Mol Gastroenterol Hepatol. 2016;2:358–368.e354.
    1. Kohi S, Sato N, Koga A. et al. KIAA1199 is induced by inflammation and enhances malignant phenotype in pancreatic cancer. Oncotarget. 2017;8:17156–17163.
    1. Zhang D, Zhao L, Shen Q. et al. Down-regulation of KIAA1199/CEMIP by miR-216a suppresses tumor invasion and metastasis in colorectal cancer. Int J Cancer. 2017;140:2298–2309.
    1. Jami MS, Hou J, Liu M. et al. Functional proteomic analysis reveals the involvement of KIAA1199 in breast cancer growth, motility and invasiveness. BMC cancer. 2014;14:194.
    1. Koga A, Sato N, Kohi S. et al. KIAA1199/CEMIP/HYBID overexpression predicts poor prognosis in pancreatic ductal adenocarcinoma. Pancreatology. 2017;17:115–22.
    1. Suh HN, Jun S, Oh AY. et al. Identification of KIAA1199 as a biomarker for pancreatic intraepithelial neoplasia. Sci Rep. 2016;6:38273.
    1. Koh Y, Kim D, Jung WJ. et al. Revealing genomic profile that underlies tropism of myeloma cells using whole exome sequencing. Int J Genomics. 2015;2015:675379.
    1. Chanthammachat P, Promwikorn W, Pruegsanusak K. et al. Comparative proteomic analysis of oral squamous cell carcinoma and adjacent non-tumour tissue from thailand. Arch Oral Biol. 2013;58:1677–1685.
    1. Matsuzaki S, Tanaka F, Mimori K. et al. Clinicopathologic significance of KIAA1199 overexpression in human gastric cancer. Ann Surg Oncol. 2009;16:2042–2051.
    1. Motaln H, Gruden K, Hren M. et al. Human mesenchymal stem cells exploit the immune response mediating chemokines to impact the phenotype of glioblastoma. Cell Transplant. 2012;21:1529–1545.
    1. Sabates-Bellver J, Van der Flier LG, de Palo M. et al. Transcriptome profile of human colorectal adenomas. Mol Cancer Res. 2007;5:1263–1275.
    1. Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480.
    1. Shostak K, Zhang X, Hubert P. et al. NF-κB-induced KIAA1199 promotes survival through EGFR signalling. Nat Commun. 2014;5:5232.
    1. Shostak K, Chariot A. EGFR and NF-κB: Partners in cancer. Trends Mol Med. 2015;21:385–393.
    1. Kikuchi M, Yamashita K, Waraya M. et al. Epigenetic regulation of ZEB1-RAB25/ESRP1 axis plays a critical role in phenylbutyrate treatment-resistant breast cancer. Oncotarget. 2016;7:1741–1753.
    1. Axelson H, Fredlund E, Ovenberger M. et al. Hypoxia-induced dedifferentiation of tumor cells-a mechanism behind heterogeneity and aggressiveness of solid tumors. Semin Cell Dev Biol. 2005;16:554–563.
    1. Vaupel P. Hypoxia and aggressive tumor phenotype: Implications for therapy and prognosis. Oncologist. 2008;13(Suppl 3):21–26.
    1. Evensen NA, Li Y, Kuscu C. et al. Hypoxia promotes colon cancer dissemination through up-regulation of cell migration-inducing protein (CEMIP) Oncotarget. 2015;6:20723–20739.
    1. Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev. 2016;97:186–203.
    1. Paulis YW, Huijbers EJ, van der Schaft DW. et al. CD44 enhances tumor aggressiveness by promoting tumor cell plasticity. Oncotarget. 2015;6:19634–19646.
    1. Nagaoka A, Yoshida H, Nakamura S. et al. Regulation of hyaluronan (HA) metabolism mediated by HYBID (hyaluronan-binding protein involved in HA depolymerization, KIAA1199) and HA synthases in growth factor-stimulated fibroblasts. J Biol Chem. 2015;290:30910–30923.
    1. Yamamoto H, Tobisawa Y, Inubushi T. et al. A mammalian homolog of the Zebrafish Transmembrane Protein 2 (TMEM2) is the long-sought-after cell surface hyaluronidase. J Biol Chem. 2017;292:7304–7313.
    1. Ruedel A, Dietrich P, Schubert T. et al. Expression and function of microrna-188-5p in activated rheumatoid arthritis synovial fibroblasts. Int J Clin Exp Pathol. 2015;8:4953–4962.
    1. Shinohara T, Izawa T, Mino-Oka A. et al. Hyaluronan metabolism in overloaded temporomandibular joint. J Oral Rehabil. 2016;43:921–928.
    1. Krishnamurthy VK, Stout AJ, Sapp MC. et al. Dysregulation of hyaluronan homeostasis during aortic valve disease. Matrix Biol. 2016 [Epub ahead of print]
    1. Schmaus A, Bauer J, Sleeman JP. Sugars in the microenvironment: The sticky problem of HA turnover in tumors. Cancer Metastasis Rev. 2014;33:1059–1079.
    1. Nykopp TK, Pasonen-Seppanen S, Tammi MI. et al. Decreased hyaluronidase 1 expression is associated with early disease recurrence in human endometrial cancer. Gynecol Oncol. 2015;137:152–159.
    1. Toole BP. Hyaluronan: From extracellular glue to pericellular cue. Nat Rev Cancer. 2004;4:528–539.

Source: PubMed

3
订阅