Cancer Letters

Cancer Letters

Volume 412, 1 January 2018, Pages 143-154
Cancer Letters

Original Article
Pancreatic stellate cells reorganize matrix components and lead pancreatic cancer invasion via the function of Endo180

https://doi.org/10.1016/j.canlet.2017.10.010Get rights and content

Highlights

  • Pancreatic stellate cells (PSCs) lead pancreatic cancer cells (PCCs) invasion by extracellular matrix (ECM) remodeling.

  • Endo180 is related with invasive ability of leading PSCs via phosphorylation of myosin light chain 2 (MLC2).

  • Inhibition of Endo180 in PSCs suppressed the ability of ECM remodeling and consequently suppressed co-cultured PCCs invasion.

Abstract

Specific cell populations leading the local invasion of cancer are called “leading cells”. However, the underlying mechanisms are unclear. Here, we identified leading cells in pancreatic cancer and determined how these cells lead and promote cancer cell invasion in the extracellular matrix (ECM). Using three-dimensional matrix remodeling assay, we found that pancreatic stellate cells (PSCs) frequently invaded the collagen matrix with pancreatic cancer cells (PCCs), which invaded behind the invading PSCs. In addition, invading PSCs changed the alignment of collagen fibers, resulting in ECM remodeling and an increase in the parallel fibers along the direction of invading PSCs. Endo180 expression was higher in PSCs than in PCCs, Endo180 knockdown in PSCs attenuated the invasive abilities of PSCs and co-cultured PCCs, and decreased the expression level of phosphorylated myosin light chain 2 (MLC2). In mouse models, Endo180-knockdown PSCs suppressed tumor growth and changes in collagen fiber orientation in co-transplantation with PCCs. Our findings suggest that PSCs lead the local invasion of PCCs by physically remodeling the ECM, possibly via the function of Endo180, which reconstructs the actin cell skeleton by phosphorylation of MLC2.

Introduction

Pancreatic cancer is one of the most lethal cancers, and its 5-year survival rate is <8% [1], largely owing to the difficulty in early detection, fast progression (e.g., local invasion into surrounding organs and distant metastasis), and the poor improvements in treatments. Radical resection is the only curative treatment. However, pancreatic cancer patients that undergo radical operation have a poor prognosis for distant metastasis and local recurrence. Therefore, it is necessary to elucidate the pathogenesis of pancreatic cancer and understand the mechanisms of invasion and metastasis to develop a new treatment for the improvement of such a poor prognosis.

Pancreatic cancer is characterized by excessive desmoplasia with an abundant extracellular matrix (ECM), which plays a crucial role in its aggressive behavior [2], and resistance to traditional therapies through tumor-stromal interactions [3]. The enriched desmoplastic microenvironment in the ECM is mainly produced by activated pancreatic stellate cells (PSCs). They are activated upon stimulation by various autocrine and paracrine factors. PSCs express α-smooth muscle actin (αSMA) and produce various ECM proteins [4], [5]. PSC-derived soluble factors and an abundant ECM promote the proliferation, migration, and invasion of pancreatic cancer cells (PCCs), leading to poor survival of pancreatic cancer patients [6], [7].

During cancer cell invasion, the specific cell populations leading the local invasion are known as “leading cells”. Previously, Gaggioli et al. [8] reported that cancer-associated fibroblasts lead and promote squamous cancer cell invasion through matrix metalloproteinases (MMPs) and force-dependent generation of ECM tracks. Cheung et al. [9] reported that K14-positive cells directly modify tensile strength and viscoelastic properties to regulate cell–cell adhesion or change signal transduction mechanisms independently of their mechanical properties, leading to collective invasion of human breast cancer. Brentnall [10] reported that palladin-activated fibroblasts promote PCC invasion by creating tunnels though the ECM. However, the underlying mechanisms of the phenomenon have been unclear.

Remodeling of the ECM is considered to play a major role in pancreatic cancer invasion and metastasis [11]. ECM remodeling continues in cancer tissue and causes resistance to established therapies [12], [13]. Consequently, we need to understand the mechanisms of ECM remodeling to elucidate the invasive and metastatic behaviors of PCCs.

Endo180 (also known as CD280, MRC2, and urokinase-type plasminogen activator receptor-associated protein) is a multiple C-type lectin-like domain receptor that binds to extracellular collagens via its fibronectin type II domain [14]. It is essential for cellular uptake of collagen in fibroblasts and plays a physiological role in mediating collagen matrix remodeling during tissue development [14]. Furthermore, Endo180 induces Rho-ROCK-myosin light chain 2 (MLC2)-based contractile signals and promotes adhesion disassembly [15]. Endo180 is highly expressed in fibroblasts. Recently, Endo180 was found to be expressed in not only fibroblasts but also several cancers [16], [17], [18], [19]. Moreover, Endo180 is related to cancer cell survival in prostate cancer [18] and breast cancer [20]. These data suggest that Endo180 plays an important role in ECM remodeling and cancer progression. However, the mechanisms by which Endo180 contributes to the ECM remodeling of pancreatic cancer are not understood.

In the present study, we used in vitro three-dimensional (3D) matrix remodeling assay to identify the leading cells in pancreatic cancer and determine how these cells lead and promote cancer cell invasion in the ECM.

Section snippets

Materials and methods

Detailed information is provided in the Supplementary Materials and methods.

PSCs lead and promote local invasion of PCCs

To establish an in vitro model similar to the in vivo microenvironment of pancreatic cancer, we performed a 3D matrix remodeling assay in which PCCs and/or PSCs were embedded in the center of the collagen matrix (Fig. 1A). The movements of PSCs and PCCs were investigated in the surrounding collagen matrix, and we evaluated the effect of the physical behavior of PSCs on the invasive behavior of PCCs in the collagen matrix.

In the 3D matrix remodeling assay, PSC1 and SUIT2 more frequently invaded

Discussions

An abundant ECM is a major characteristic of pancreatic cancer. To reproduce the ECM, we used a 3D matrix remodeling assay and found that PSCs invaded into the collagen matrix and cancer cells frequently invaded following the invading PSCs in co-cultures of PCCs and PSCs. These observations suggest that PCCs and PSCs enhance the invasive abilities of each other by tumor-stromal (cancer cell-PSC) interactions as reported previously [2], [3], [23], [36], [37]. Furthermore, invading cancer cells

Acknowledgements

The authors thank E. Manbe, S. Sadatomi (Department of Surgery and Oncology, Kyushu University Hospital), and members of the Research Support Center and Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, for their expert technical assistance.

References (50)

  • M. Erkan et al.

    StellaTUM: current consensus and discussion on pancreatic stellate cell research

    Gut

    (2012)
  • A. Neesse et al.

    Stromal biology and therapy in pancreatic cancer: a changing paradigm

    Gut

    (2015)
  • E. Schneider et al.

    Identification of mediators stimulating proliferation and matrix synthesis of rat pancreatic stellate cells

    Am. J. Physiol. Cell Physiol.

    (2001)
  • P. Mews et al.

    Pancreatic stellate cells respond to inflammatory cytokines: potential role in chronic pancreatitis

    Gut

    (2002)
  • H. Habisch et al.

    Interaction of stellate cells with pancreatic carcinoma cells

    Cancers (Basel)

    (2010)
  • A. Wehr et al.

    Analysis of the human pancreatic stellate cell secreted proteome

    Pancreas

    (2011)
  • C. Gaggioli et al.

    Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells

    Nat. Cell Biol.

    (2007)
  • T.A. Brentnall

    Arousal of cancer-associated stromal fibroblasts: palladin-activated fibroblasts promote tumor invasion

    Cell Adhes. Migr.

    (2012)
  • T.R. Cox et al.

    Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer

    Dis. Model. Mech.

    (2011)
  • K.P. Olive et al.

    Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer

    Science

    (2009)
  • N.S. Nagathihalli et al.

    Signal transducer and activator of transcription 3, mediated remodeling of the tumor microenvironment results in enhanced tumor drug delivery in a mouse model of pancreatic cancer

    Gastroenterology

    (2015)
  • D. Wienke et al.

    Identification and characterization of the endocytic transmembrane glycoprotein Endo180 as a novel collagen receptor

    Mol. Biol. Cell.

    (2003)
  • J. Sturge et al.

    Endosomes generate localized Rho-ROCK-MLC2-based contractile signals via Endo 180 to promote adhesion disassembly

    J. Cell Biol.

    (2006)
  • I.J. Huijbers et al.

    A role for fibrillar collagen deposition and the collagen internalization receptor endo180 in glioma invasion

    PLoS One

    (2010)
  • D. Wienke et al.

    The collagen receptor Endo180 (CD280) Is expressed on basal-like breast tumor cells and promotes tumor growth in vivo

    Cancer Res.

    (2007)
  • Cited by (31)

    • Targeting Pin1 renders pancreatic cancer eradicable by synergizing with immunochemotherapy

      2021, Cell
      Citation Excerpt :

      Thus, Pin1 is overexpressed in cancer cells and CAFs in human PDAC and correlates well with the desmoplastic and immunosuppressive TME and poor survival. To determine the functional significance of Pin1 overexpression in CAFs in human PDAC tumors, we derived primary CAFs from two different PDAC patients (Koikawa et al., 2018a, 2018b) and inhibited their Pin1 function using Pin1i, genetic knockdown (KD) or CRISPR knockout (KO). Both Pin1 inhibitors dose-dependently reduced Pin1 and its many substrate oncoproteins in CAFs (Figures 3A and S3C), with ATRA and ATO synergistically targeting and degrading Pin1, as shown (Kozono et al., 2018; Wei et al., 2015).

    • Bone marrow-derived macrophages converted into cancer-associated fibroblast-like cells promote pancreatic cancer progression

      2021, Cancer Letters
      Citation Excerpt :

      In newborn KPC recipient pancreas, BM-derived GFP+ cells were more frequently engrafted at the invasive front and central lesions of PDAC compared with normal acini (Fig. 1B). The border between the tumor and the acini can be histopathologically divided into two distinct regions, the invasive front and the non-invasive front, according to our previous report [14,20]. As shown in Fig. 1C, GFP+ cells were detected in newborn KPC recipient pancreas, BM, PB, and liver upon fluorescence-activated cell sorting (FACS) analysis.

    View all citing articles on Scopus
    View full text