CD133 as a regulator of cancer metastasis through the cancer stem cells

https://doi.org/10.1016/j.biocel.2018.10.013Get rights and content

Abstract

Cancer stem cells are the cancer cells that have abilities to self-renew, differentiate into defined progenies, and initiate and maintain tumor growth. They also contribute to cancer metastasis and therapeutic resistance, both of which are the major causes of cancer mortality. Among the reported makers of the cancer stem cells, CD133 is the most well-known marker for isolating and studying cancer stem cells in different types of cancer. The CD133high population of cancer cells are not only capable of self-renewal, proliferation, but also highly metastatic and resistant to therapy. Despite very limited information on physiological functions of CD133, many ongoing studies are aimed to reveal the mechanisms that CD133 utilizes to modulate cancer dissemination and drug resistance with a long-term goal for bringing down the number of cancer deaths. In this review, in addition to the regulation of CD133, and its involvement in cancer initiation, and development, the recent updates on how CD133 modulates cancer dissemination, and therapeutic resistance are provided. The key signaling pathways that are upstream or downstream of CD133 during these processes are summarized. A comprehensive understanding of CD133-mediated cancer initiation, development, and dissemination through its pivotal role in cancer stem cells will offer new strategies in cancer therapy.

Introduction

CD133 is a glycosylated transmembrane protein, encoded by PROM1 “Prominin-1. It has five transmembrane domains across the plasma membrane with an extracellular NH2 terminus and an intracellular COOH terminus. The physiological functions of CD133, so far, are mainly reported in retinal development. PROM1 mutations are harbored in the populations suffering from retinitis pigmentosa, macular degeneration and cone-rod retinal dystrophy (Maw et al., 2000; Michaelides et al., 2010; Permanyer et al., 2010; Yang et al., 2008; Zhang et al., 2007). In addition, reduced adhesion abilities and increased cell damages were detected in the peripheral endothelial cells that harbor CD133 missense mutation (Arrigoni et al., 2011).

CD133 is originally discovered in the human hematopoietic stem and progenitor cells (Miraglia et al., 1997; Yin et al., 1997). Accumulating evidence indicated a presence of the high protein levels of CD133 in numerous types of cancer. The highly expressed CD133 predicts poor outcomes of cancer patients of ovarian cancer, colorectal cancer, prostate cancer, rectal cancer, lung cancer, and glioblastoma (Horst et al., 2009b; Merlos-Suarez et al., 2011; Ong et al., 2010; Silva et al., 2011; Artells et al., 2010; Hurt et al., 2008; Saigusa et al., 2009; Zeppernick et al., 2008; Zhang et al., 2008; Alamgeer et al., 2013; Huang et al., 2015; Wu et al., 2014). This is because cancer cells that express high levels of CD133 are more metastatic and resistant to chemotherapy and radiation therapy. Given that CD133+ cells are capable of self-renewal, proliferation and differentiation into different types of cells (Hemmati et al., 2003; Singh et al., 2003, 2004; Yin et al., 1997), known as stem cell properties, CD133+ cancer cells are cancer stem cells (CSCs). In addition to CD133, other general cancer stem cell markers include CD44 and aldehyde dehydrogenase1A1 (ALDH1A1). Heterogeneous populations of the CSCs are present among different types of cancer according to their protein expression profiles. For example, pancreatic cancer stem cells express high levels of CD133, CD44, CD24, epithelial-specific antigen (ESA), ALDH1A1, CXCR4, DCLK-1 and BMI-1, while lung cancer stem cells have increased expression of ALDH1A1, ABCG2, CD90, CD117 and epithelial cellular adhesion molecule (EpCAM) (Hardavella et al., 2016; Proctor et al., 2013; Rao and Mohammed, 2015; Wang et al., 2014).

The CD133 expression is regulated by Notch, p53, hypoxia-inducing factor (HIF) and signal transducer and activator of transcription 3 (STAT3) in cancer (Fig. 1). It has been demonstrated that the intracellular domain of Notch 1 directly bound to the RBP-Jκ site of the 5′ promoter region of PROM1 to regulate CD133 transcription (Konishi et al., 2016). Knockdown of Notch1 or treatment of Notch inhibitors decreased CD133 expression in cultured gastric cancer and melanoma cells (Konishi et al., 2016; Kumar et al., 2016). There are 5 different promoters, including promoter 1 (P1) to promoter 5 (P5) in the 5´ untranslated region of CD133 for alternatively splicing variants. HIF increased the promoter activity of PROM1 through its direct binding to the P5 region of PROM1 where it interacted with ETS transcription factors such as Elk1 (Ohnishi et al., 2013). Recently, it has been reported that STAT3 activated by IL-6 can turn on the PROM1 gene through upregulation of HIF transcription in liver cancer cells (Won et al., 2015). In human lung cancer cells cultured at a hypoxia condition, binding of OCT4 and SOX2 to the P1 region of PROM1 was required for HIF-induced CD133 expression (Iida et al., 2012), revealing another mechanism that HIF modulates CD133 expression in addition to the P5 region of PROM1. Transcription factor p53 negatively regulates mRNA and protein levels of CD133 by directly binding to the P5 region of PROM1, and subsequent recruitment and activation of histone deacetylase 1 (HDAC1) (Park et al., 2015). Activation of HDAC1 removed the acetyl groups from lysine residues of the chromosome, and subsequently increased the binding between the histones and DNA, thus preventing transcription of PROM1.

This review provides the current updates on how CD133 regulates different stages of cancer development, including initiation, progression and advancement (metastasis) especially through transcription factors and cell signaling. In addition, recent findings on the mechanisms that CD133 utilizes to enhance cancer cells resistances to therapeutic treatments are also summarized for offering some insights into a potential modulation on CD133 and its mediated signaling for a therapeutic development purpose.

Section snippets

CD133 in cancer initiation

Cancer-initiating cells possess tumor-initiating capacity which is one of the characteristics of CSCs. CSCs are believed to be a major source of cancer-initiating cells during cancer onset. A major body of evidence has demonstrated that the isolated CD133+ cancer cells from patients are capable of forming cancers in immune-comprised xenograft mice, implicating the involvement of CSCs in cancer initiation. When using the renal capsule transplantation in immunodeficient mice to identify human

CD133 in cancer development and progression

In a xenograft mouse model, the size of tumor derived from the high CD133+ HEK293 cells is dramatically larger than that from the low CD133+ HEK293 cells (Canis et al., 2013), suggesting that a role of CD133 in regulating cell growth during cancer development. Overexpression of CD133 increased cell proliferation, cell cycle progression and telomerase activity in pancreatic cancer AsPC-1 cells (Weng et al., 2016), suggesting that CD133 promotes tumor progression through upregulation of cell

CD133 and its signaling in cancer metastasis

A little more than a decade ago after discovering CD133 as a marker of brain tumor stem cells, accuVmulating evidence suggested that CD133 modulates cancer cell invasion, metastasis and drug resistance in many types of cancer (Fig. 3).

In metastatic ovarian cancer, increased mRNA expression of CD133 is regulated by transcription factor ARID3B. Knockdown of CD133 in ARID3B overexpressed cancer cells leads to quicker tumor-caused deaths in the xenograft mice as compared to these of CD133+ARID3B+

Potential of targeting CD133 in cancer therapy

Several lines of evidence have suggested CD133 as a prognostic marker in many types of cancers including breast cancer, lung cancer, gastric cancer, colorectal cancer and so on (Alamgeer et al., 2013; Horst et al., 2009b; Ishigami et al., 2010; Wu et al., 2014; Xia, 2017). In addition to cancer initiation, development, and metastasis, CD133 also enhances therapeutic resistance including chemo drugs and radiation. Overexpression of CD133 in a head and neck squamous cell carcinoma (HNSCC) cell

Summary

Tumor metastasis and therapeutic resistance account for the majority of cancer-related deaths. Both properties are enhanced in a small population of CD133high cancer cells, known as cancer stem cells with abilities of self-renewal, tumor initiation, and pluripotency. Therefore, targeting cancer stem cells, such as CD133high cell populations remains one of top interest to combat fatalities of cancer patients. Elevated expression of CD133 resulted in increased tumor-initiating ability, tumor

Funding

This work was supported by the NIH Grant G12MD007590 and by the 2017 AACR-Bayer Innovation and Discovery Grant 17-80-44-LIOU.

Competing interests

The author declares no competing interests on publication of this article.

Ethics approval and consent to participate

All animal experiments were approved by the Atlanta University Center IACUC committee (protocol #: 17-20) and performed in accordance with relevant institutional and national guidelines and regulations.

References (91)

  • C. Liu et al.

    The interaction between Cancer stem cell marker CD133 and src protein promotes focal adhesion kinase (FAK) phosphorylation and cell migration

    J. Biol. Chem.

    (2016)
  • A.B. Mak et al.

    Regulation of CD133 by HDAC6 promotes beta-catenin signaling to suppress cancer cell differentiation

    Cell Rep.

    (2012)
  • A. Merlos-Suarez et al.

    The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse

    Cell Stem Cell

    (2011)
  • S. Miraglia et al.

    A novel five-transmembrane hematopoietic stem cell antigen: isolation, characterization, and molecular cloning

    Blood

    (1997)
  • C.W. Ong et al.

    CD133 expression predicts for non-response to chemotherapy in colorectal cancer

    Mod. Pathol.

    (2010)
  • G. Rappa et al.

    Wnt interaction and extracellular release of prominin-1/CD133 in human malignant melanoma cells

    Exp. Cell Res.

    (2013)
  • W.S. Song et al.

    Sox2, a stemness gene, regulates tumor-initiating and drug-resistant properties in CD133-positive glioblastoma stem cells

    J. Chin. Med. Assoc.

    (2016)
  • S.K. Swaminathan et al.

    CD133-targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer

    J. Control. Release

    (2013)
  • M.R. Webster et al.

    The Wnts of change: how Wnts regulate phenotype switching in melanoma

    Biochim. Biophys. Acta

    (2015)
  • C.B. Westphalen et al.

    Dclk1 defines quiescent pancreatic progenitors that promote injury-induced regeneration and tumorigenesis

    Cell Stem Cell

    (2016)
  • A.H. Yin et al.

    AC133, a novel marker for human hematopoietic stem and progenitor cells

    Blood

    (1997)
  • M. Alamgeer et al.

    The prognostic significance of aldehyde dehydrogenase 1A1 (ALDH1A1) and CD133 expression in early stage non-small cell lung cancer

    Thorax

    (2013)
  • J.M. Angelastro et al.

    Overexpression of CD133 promotes drug resistance in C6 glioma cells

    Mol. Cancer Res.

    (2010)
  • F.I. Arrigoni et al.

    Extended extraocular phenotype of PROM1 mutation in kindreds with known autosomal dominant macular dystrophy

    Eur. J. Hum. Genet.

    (2011)
  • D. Beier et al.

    CD133(+) and CD133(-) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles

    Cancer Res.

    (2007)
  • A. Brossa et al.

    Role of CD133 molecule in wnt response and renal repair

    Stem Cells Transl. Med.

    (2018)
  • M. Canis et al.

    CD133 induces tumour-initiating properties in HEK293 cells

    Tumour Biol.

    (2013)
  • K.L. Chen et al.

    Highly enriched CD133(+)CD44(+) stem-like cells with CD133(+)CD44(high) metastatic subset in HCT116 colon cancer cells

    Clin. Exp. Metastasis

    (2011)
  • Y.S. Chen et al.

    CD133/Src axis mediates tumor initiating property and epithelial-mesenchymal transition of head and neck cancer

    PLoS One

    (2011)
  • M.D. Curley et al.

    CD133 expression defines a tumor initiating cell population in primary human ovarian cancer

    Stem Cells

    (2009)
  • K.E. Delgiorno et al.

    Identification and manipulation of biliary metaplasia in pancreatic tumors

    Gastroenterology

    (2014)
  • Y. Deng et al.

    Celecoxib downregulates CD133 expression through inhibition of the Wnt signaling pathway in colon cancer cells

    Cancer Invest.

    (2013)
  • Q. Ding et al.

    CD133 facilitates epithelial-mesenchymal transition through interaction with the ERK pathway in pancreatic cancer metastasis

    Mol. Cancer

    (2014)
  • C.L. Eaton et al.

    Evaluation of the frequency of putative prostate cancer stem cells in primary and metastatic prostate cancer

    Prostate

    (2010)
  • X. Fan et al.

    NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts

    Stem Cells

    (2010)
  • K.C. Feng et al.

    Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced Cholangiocarcinoma

    J. Hematol. Oncol.

    (2017)
  • N. Garg et al.

    CD133(+) brain tumor-initiating cells are dependent on STAT3 signaling to drive medulloblastoma recurrence

    Oncogene

    (2017)
  • R. Handgretinger et al.

    CD133-positive hematopoietic stem cells: from biology to medicine

    Adv. Exp. Med. Biol.

    (2013)
  • G. Hardavella et al.

    Lung cancer stem cells-characteristics, phenotype

    Transl. Lung Cancer Res.

    (2016)
  • H.D. Hemmati et al.

    Cancerous stem cells can arise from pediatric brain tumors

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • D. Horst et al.

    Prognostic significance of the cancer stem cell markers CD133, CD44, and CD166 in colorectal cancer

    Cancer Invest.

    (2009)
  • Y. Hou et al.

    The critical role of CD133(+)CD44(+/high) tumor cells in hematogenous metastasis of liver cancers

    Cell Res.

    (2012)
  • M. Huang et al.

    High CD133 expression in the nucleus and cytoplasm predicts poor prognosis in non-small cell lung cancer

    Dis. Markers

    (2015)
  • E.M. Hurt et al.

    CD44+ CD24(-) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis

    Br. J. Cancer

    (2008)
  • H. Iida et al.

    Hypoxia induces CD133 expression in human lung cancer cells by up-regulation of OCT3/4 and SOX2

    Int. J. Oncol.

    (2012)
  • Cited by (0)

    View full text