Elsevier

Seminars in Cancer Biology

Volume 29, December 2014, Pages 51-58
Seminars in Cancer Biology

Review
The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition

https://doi.org/10.1016/j.semcancer.2014.08.003Get rights and content

Abstract

Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78 kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.

Introduction

Tumors may be considered as caricatures of the process of normal embryonic development whereby oncogeny recapitulates ontogeny in an inappropriate spatiotemporal context [1], [2]. Specifically, the subversion and corruption of embryonic signaling pathways such as Wntβ-catenin, Notch/Cbf-1, Hedgehog/Gli and Nodal/CR-1 may be instrumental as drivers in the initiation and/or progression of multiple types of cancer especially if these pathways are operative in CSCs (the cells thought to propagate the tumor) or transit amplifying (TA) progenitor populations [2], [3]. The loss of appropriate genetic or epigenetic regulatory mechanisms that might occur in normal adult somatic tissue stem cells (SCs), TA cells or the surrounding niche cell populations is a likely contributor to the alteration in expression and/or aberrant activation of these embryonic signaling pathways observed in tumors. The consequences of these alterations may then lead to a disruption in the cell-cell communication between different tissue compartments (epithelial and stromal) and a loss in normal tissue architecture as mediated by the processes of EMT and mesenchymal-epithelial transition (MET).

The normal tissue microenvironment also has a significant influence on the suppression, initiation, and progression of tumor cells. For example, the embryonic microenvironment or the adult stem cell niche can reprogram tumor cells to acquire a more normal cellular lineage restriction and to differentiate [4], [5]. Reciprocally, the tumor microenvironment that consists of myeloid suppressor cells, mesenchymal stem cells which are derived from the bone marrow or surrounding cancer-associated fibroblasts (CAFs) can directly or indirectly through secreted factors reprogram SCs and induced-pluripotent stem cells (iPSCs) to acquire properties of CSCs or tumor initiating cells (TICs) [6], [7]. Identification of these factors that are expressed in cancer cells or by the surrounding niche compartment may provide unique drug targets for cancer therapy.

In this review, we discuss the novel biological properties of the embryonic gene CR-1 and the molecular signaling pathways that are regulated by CR-1 which may contribute to its pro-tumorigenic role in various types of cancer. The expression of CR-1 in potential CSCs or TICs suggests that CR-1 coupled with its capacity to facilitate EMT could prove to be an efficacious therapeutic target for the clinical management of malignant disease.

Section snippets

Structure and mechanisms regulating expression of Cripto-1

Cripto-1/TDGF-1 is the original member of the epidermal growth factor (EGF)-Cripto-1-FRL-1-Cryptic (CFC) family of vertebrate signaling molecules. It was initially isolated from human (CR-1) NTERA-2 and mouse (Cr-1) F9 undifferentiated teratocarcinoma cells [8]. Structurally, Cripto-1 is a cell membrane-associated protein containing signal sequences for extracellular secretion, a modified EGF-like domain, a conserved cysteine-rich domain (CFC-motif) and a short hydrophobic carboxy-terminus,

Role of Cripto-1 in embryogenesis and stem cell maintenance

During embryonic development in the mouse, Cr-1 is initially detected prior to gastrulation, in the inner cell mass and in extraembryonic trophoblast cells in the 4-day blastocyst. The highest Cr-1 expression is detected in epiblast cells undergoing EMT that are migrating and that give rise to the mesoderm and endoderm. Cr-1 and Cryptic signaling are involved in regulating the formation of the primitive streak, patterning of the anterior/posterior axis, specification of mesoderm and endoderm

Cripto-1 interacting partners in cellular signaling

Cripto-1 has multiple binding partners and can modulate a variety of intracellular signaling pathways implicated in embryogenesis and oncogenic transformation (Fig. 1). During embryogenesis, Cripto-1 functions primarily as a coreceptor for the TGF-β family ligands Nodal and growth and differentiation factors (GDFs) 1 and 3, leading to the activation of type I (Alk4/Alk7) serine-threonine kinase receptors and the Activin type II receptor complex, which triggers both the phosphorylation and

Cripto-1: EMT and cancer stem cells

Broadly defined, CSCs are subsets of cells in various tumors that exhibit enhanced tumorigenicity in experimental settings and which are able to reestablish the cellular heterogeneity of the original tumor [61], [62]. CSCs, also known as tumor initiating cells, share several characteristics that have been associated with normal tissue SCs [63]. CSCs were first described in tumors of hematopoietic origin [61], [62] and have now been identified in several types of solid tumors, including cancers

Cripto-1 in transformation, migration, invasion and angiogenesis

Reactivation of certain signaling pathways that are crucial during embryonic development might induce cellular transformation and tumor progression in adult tissues [95]. CR-1 is a typical example of an embryonic gene that is re-expressed during tumorigenesis, functioning as an oncogene and driving cellular proliferation, migration, and invasion, as well as stimulating tumor angiogenesis in vitro and in vivo [30], [96]. CR-1 was first demonstrated to induce cellular transformation in vitro in

Expression of Cripto-1 in human carcinomas and premalignant lesions

As previously discussed in this review, CR-1 is not significantly expressed at significant levels in adult somatic tissues, with the possible exception of the tissue SC compartment, and its re-expression can be observed during oncogenic transformation. In addition to functioning as an oncogene in vitro and in vivo, CR-1 overexpression is detected at the mRNA and protein levels in a wide variety of solid human tumors of non-neuronal origin, including those of the reproductive and

Cripto-1 as a therapeutic target in human cancer

Due to its intimate involvement in processes such as oncogenesis and EMT and minimal expression in adult tissues, CR-1 may be considered as an attractive target for therapeutic intervention. In particular, the association of CR-1 with CSCs is intriguing as this population of cells is intrinsically resistant to standard chemotherapy and radiotherapy [105]. Current strategies that can effectively target and neutralize the potential oncogenic effects of CR-1 include the use of antisense (AS)

Conclusion and perspectives

The abnormal spatial and temporal reexpression of embryonic signaling genes at different stages of tumor development in a variety of human cancers is now a well-recognized fact. In particular, the subversion of these key regulatory genes in CSCs or transit amplifying progenitor cells in human cancers may be extremely deleterious for restricting tumor progression and for preventing the re-emergence of secondary cancer following the use of primary chemo- and/or radiotherapy. Therefore, the

References (118)

  • N.C. Boles et al.

    NPTX1 regulates neural lineage specification from human pluripotent stem cells

    Cell Rep

    (2014)
  • K. Miharada et al.

    Cripto regulates hematopoietic stem cells as a hypoxic-niche-related factor through cell surface receptor GRP78

    Cell Stem Cell

    (2011)
  • B.T. Spike et al.

    CRIPTO/GRP78 signaling maintains fetal and adult mammary stem cells ex vivo

    Stem Cell Rep

    (2014)
  • C. Bianco et al.

    Cripto-1: an oncofetal gene with many faces

    Curr Top Dev Biol

    (2005)
  • M. Hirota et al.

    Smad2 functions as a co-activator of canonical Wnt/beta-catenin signaling pathway independent of Smad4 through histone acetyltransferase activity of p300

    Cell Signal

    (2008)
  • T. Nagaoka et al.

    Cripto-1 enhances the canonical Wnt/beta-catenin signaling pathway by binding to LRP5 and LRP6 co-receptors

    Cell Signal

    (2013)
  • T. Uchida et al.

    A novel epidermal growth factor-like molecule containing two follistatin modules stimulates tyrosine phosphorylation of erbB-4 in MKN28 gastric cancer cells

    Biochem Biophys Res Commun

    (1999)
  • C. Bianco et al.

    Cripto-1 indirectly stimulates the tyrosine phosphorylation of erb B-4 through a novel receptor

    J Biol Chem

    (1999)
  • C. Bianco et al.

    Regulation of Cripto-1 signaling and biological activity by caveolin-1 in mammary epithelial cells

    Am J Pathol

    (2008)
  • S.C. Bendall et al.

    An enhanced mass spectrometry approach reveals human embryonic stem cell growth factors in culture

    Mol Cell Proteomics

    (2009)
  • P.C. Gray et al.

    Cripto/GRP78 modulation of the TGF-beta pathway in development and oncogenesis

    FEBS Lett

    (2012)
  • C.F. Kim et al.

    Identification of bronchioalveolar stem cells in normal lung and lung cancer

    Cell

    (2005)
  • P.C. Hermann et al.

    Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer

    Cell Stem Cell

    (2007)
  • T. Oskarsson et al.

    Metastatic stem cells: sources, niches, and vital pathways

    Cell Stem Cell

    (2014)
  • B.T. Spike et al.

    A mammary stem cell population identified and characterized in late embryogenesis reveals similarities to human breast cancer

    Cell Stem Cell

    (2012)
  • S.A. Mani et al.

    The epithelial-mesenchymal transition generates cells with properties of stem cells

    Cell

    (2008)
  • C. Scheel et al.

    Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links

    Sem Cancer Biol

    (2012)
  • C. Wechselberger et al.

    Cripto-1 enhances migration and branching morphogenesis of mouse mammary epithelial cells

    Exp Cell Res

    (2001)
  • E. Lonardo et al.

    Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy

    Cell Stem Cell

    (2011)
  • C. Scheel et al.

    Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast

    Cell

    (2011)
  • M.C. Rangel et al.

    Role of Cripto-1 during epithelial-to-mesenchymal transition in development and cancer

    Am J Pathol

    (2012)
  • Y. Sun et al.

    Overexpression of human Cripto-1 in transgenic mice delays mammary gland development and differentiation and induces mammary tumorigenesis

    Am J Pathol

    (2005)
  • G.B. Pierce et al.

    Tumors as caricatures of the process of tissue renewal: prospects for therapy by directing differentiation

    Cancer Res

    (1988)
  • R.N. Johnston et al.

    The origin of the cancer cell: oncogeny reverses phylogeny

    Biochem Cell Biol

    (1992)
  • O. Dreesen et al.

    Signaling pathways in cancer and embryonic stem cells

    Stem Cell Rev

    (2007)
  • D.E. Abbott et al.

    The epigenetic influence of tumor and embryonic microenvironments: how different are they?

    Cancer Microenviron

    (2008)
  • L. Chen et al.

    A model of cancer stem cells derived from mouse induced pluripotent stem cells

    PLoS ONE

    (2012)
  • M. Nishi et al.

    Induction of cells with cancer stem cell properties from nontumorigenic human mammary epithelial cells by defined reprogramming factors

    Oncogene

    (2014)
  • A. Ciccodicola et al.

    Molecular characterization of a gene of the ‘EGF family’ expressed in undifferentiated human NTERA2 teratocarcinoma cells

    EMBO J

    (1989)
  • R. Dono et al.

    The murine cripto gene: expression during mesoderm induction and early heart morphogenesis

    Development

    (1993)
  • G. Minchiotti et al.

    Structure-function analysis of the EGF-CFC family member Cripto identifies residues essential for nodal signalling

    Development

    (2001)
  • M.M. Shen et al.

    A differential display strategy identifies Cryptic, a novel EGF-related gene expressed in the axial and lateral mesoderm during mouse gastrulation

    Development

    (1997)
  • Y.T. Yan et al.

    Dual roles of Cripto as a ligand and coreceptor in the nodal signaling pathway

    Mol Cell Biol

    (2002)
  • M. Mancino et al.

    Regulation of human Cripto-1 gene expression by TGF-beta1 and BMP-4 in embryonal and colon cancer cells

    J Cell Physiol

    (2008)
  • M. Morkel et al.

    Beta-catenin regulates Cripto- and Wnt3-dependent gene expression programs in mouse axis and mesoderm formation

    Development

    (2003)
  • A.N. Behrens et al.

    Nkx2-5 regulates Tdgf1 (Cripto) early during cardiac development

    J Clin Exp Cardiol

    (2012)
  • C. Bianco et al.

    Regulation of human Cripto-1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells

    J Cell Physiol

    (2013)
  • Y. Zhang et al.

    Spatially restricted translation of the xCR1 mRNA in Xenopus embryos

    Mol Cell Biol

    (2009)
  • F. Chen et al.

    MiR-15a-16 represses Cripto and inhibits NSCLC cell progression

    Mol Cell Biochem

    (2014)
  • C. Meno et al.

    Two closely-related left-right asymmetrically expressed genes, lefty-1 and lefty-2: their distinct expression domains, chromosomal linkage and direct neuralizing activity in Xenopus embryos

    Genes Cells

    (1997)
  • Cited by (81)

    • Maternal Cripto is critical for proper development of the mouse placenta and the placental vasculature

      2021, Placenta
      Citation Excerpt :

      In addition to being a co-receptor in the TGF-β signaling pathway, Cripto can activate Smad-independent signaling pathways such as PI3K/Akt and MAPK and facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 signaling pathways [15]. Through these various mechanisms, Cripto is implicated in: early embryogenesis, embryonic stem cell maintenance, facilitating epithelial-mesenchymal transitions and significantly enhancing tumor cell migration, invasion and angiogenesis [15]. Some studies have suggested that Cripto may also have a role in female reproduction and pregnancy maintenance.

    View all citing articles on Scopus
    View full text