Cancer Letters

Cancer Letters

Volume 379, Issue 1, 28 August 2016, Pages 49-59
Cancer Letters

Original Articles
Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-β pathways

https://doi.org/10.1016/j.canlet.2016.05.022Get rights and content

Highlights

Abstract

Fibroblasts are rich in the surrounding microenvironment of hepatocellular carcinoma (HCC) because most HCCs occur in fibrotic or cirrhotic livers. However, the role of cancer-associated fibroblasts (CAFs) in HCC metastasis remains obscure. Here, we reported that CAFs promote the migration and invasion of HCC cells in vitro and facilitate the HCC metastasis to the bone, brain and lung in NOD/SCID mice. The RayBio human chemokine antibody array revealed that CAFs secret higher levels of CCL2, CCL5, CCL7 and CXCL16 than peri-tumor fibroblasts. CCL2 and CCL5 increase the migration but not the invasion of HCC cells, while CCL7 and CXCL16 promote both migration and invasion of HCC cells. Moreover, CCL2 and CCL5 stimulate the activation of the hedgehog (Hh) pathway, while CCL7 and CXCL16 enhance the activity of the transforming growth factor-β (TGF-β) pathway in HCC cells. The neutralizing antibodies of chemokines notably attenuate the effect of CAFs on HCC metastasis and compromised the activation of Hh and TGF-β pathways in HCC cells. In summary, CAF-secreted CCL2, CCL5, CCL7 and CXCL16 promote HCC metastasis through the coordinate activation of Hh and TGF-β pathways in HCC cells.

Introduction

The tumor microenvironment is a complicated mixture of tumor cells within the extracellular matrix (ECM), including diffusible growth factors, cytokines, and various types of stromal cells. A specialized group of fibroblasts called cancer-associated fibroblasts (CAFs) is believed to actively participate in the metastasis and invasion of tumor cells [1], [2]. Evidence from clinical and epidemiological studies has shown a strong association between CAFs and poor prognosis in several types of cancer, including HCC [3], [4].

Although CAFs have a high degree of heterogeneity due to their various origins, all of them express certain mesenchymal markers including α-SMA, Vimentin, and FSP-1. However, CAFs do not express the common markers of epithelial and endothelial cells [1], [2]. In addition, CAFs are quite different from PTFs in both their molecular constitution as well as their functional impact on neighboring epithelial cells [5]. It is known that most HCCs occur in fibrotic or cirrhotic livers. The major producers of the fibrotic extracellular matrix in the liver are myofibroblasts derived from quiescent fibroblasts and hepatic stellate cells activated by chronic injury. Consistently, HCC cells are embedded in a surrounding microenvironment that is rich in fibroblasts. Previous studies have successfully isolated CAFs and PTFs from human HCC samples [4], [6]. The myofibroblast marker α-SMA is expressed at higher levels in CAFs than in normal fibroblasts [6], [7]. CAFs in HCCs also proliferated more efficiently than PTFs [6].

Previous studies have revealed that CAFs promote cancer progression and metastasis by secreting a variety of soluble factors, including inflammatory cytokines, growth factors and chemokines [6], [8]. TGF-β and hepatocyte growth factor (HGF) released by CAFs have been shown to induce the malignant transformation of human breast cells [9]. The inhibition of CAF-secreted fibroblast growth factor-2 (FGF-2) reduces angiogenesis [10]. CAF-derived HGF can active the c-Met pathway in pre-invasive DCIS and enhance the transition to invasive carcinoma in breast cancer [11]. It has been reported that CAFs also increase the proliferation and invasive property of HCC cells [12]. However, the cytokine profile of CAFs in HCC remains unclear.

As mediators of inflammation, chemokines have been reported to modulate the recruitment of inflammatory cells and, thus, modify the environment in which cancer eventually develops. Recently, several studies demonstrated that chemokines also directly affect tumor cells. CCL2 produced by CAFs stimulates cancer stem cell self-renewal of breast cancer cells [13]. CCL5 promotes tumor growth and invasion of ovarian cancer [14]. A recent study found the CXCR2/CXCL5 axis contributes to the spread of HCC by inducing the EMT through activation of the PI3K/Akt/GSK-3β/Snail pathway [15]. These findings suggest that chemokines directly promote tumor progression. Nevertheless, the role of CAF-derived chemokines in HCC progression remains elusive.

In this study, we investigated the effect of CAFs on HCC metastasis in vitro and in vivo. The differential chemokines secreted by CAFs and PTFs were also analyzed. We found that CAF-derived CCL2, CCL5, CCL7 and CXCL16 promote HCC metastasis through inducing the activation of Hh and TGF-β pathways in HCC cells.

Section snippets

Human tissues

Human liver tissues were obtained from HCC patients undergoing surgical resection at the Eastern Hepatobiliary Surgery Hospital (Shanghai, China), and all patients provided written informed consent. HCC tissues with typical macroscopic features were collected from tumor nodules and were examined with hematoxylin and eosin (H&E) staining to confirm the diagnosis. The peri-tumoral tissues without histopathologically identified tumor cells were collected from at least 5 cm away from the tumor

CAFs promote the malignant properties of HCC cells

Six paired CAF and PTF cells were isolated from HCC tissues and adjacent liver tissues and were characterized with the fibroblast markers, FSP1 and α-SMA, and the mesenchymal marker, Vimentin (Fig. S1A). Consistent with previous reports [6], CAFs expressed higher levels of fibroblast and mesenchymal markers compared with PTFs (Fig. S1B and C). In addition, CAFs also proliferated and migrated more efficiently than PTFs (Fig. S2A and B). These data confirmed that the fibroblasts surrounding HCC

Discussion

CAFs, the most common cells in the tumor stroma, play an important role in tumor-stromal cross-talk. A previous study revealed that CAFs could be involved in the proliferation, migration, and invasion of HCC cells [6]. Nevertheless, the mechanism for the involvement of CAFs in the metastasis of HCC is largely unknown. Our studies provide evidence that CAFs increase the migration and invasion abilities of HCC cells. Furthermore, CAFs markedly promote the metastasis of HCC cells in NOD/SCID mice.

Funding

This work supported by grants from the National Natural Science Foundation of China 81372675, 81572377, 81572412 and the Shanghai Science and Technology Committee for Key Project 13JC1407400.

Conflict of interest

None to declare.

References (55)

  • I. Haviv et al.

    Origin of carcinoma associated fibroblasts

    Cell Cycle

    (2009)
  • A. Mazzocca et al.

    Tumor-secreted lysophostatidic acid accelerates hepatocellular carcinoma progression by promoting differentiation of peritumoral fibroblasts in myofibroblasts

    Hepatology

    (2011)
  • H. Sugihara et al.

    Cancer-associated fibroblast-derived CXCL12 causes tumor progression in adenocarcinoma of the esophagogastric junction

    Med. Oncol

    (2015)
  • Y. Kojima et al.

    Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts

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

    (2010)
  • C. Kuperwasser et al.

    Reconstruction of functionally normal and malignant human breast tissues in mice

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

    (2004)
  • K. Pietras et al.

    Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting

    PLoS Med

    (2008)
  • C. Jedeszko et al.

    Fibroblast hepatocyte growth factor promotes invasion of human mammary ductal carcinoma in situ

    Cancer Res

    (2009)
  • A. Tsuyada et al.

    CCL2 mediates cross-talk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells

    Cancer Res

    (2012)
  • A.K. Mitra et al.

    MicroRNAs reprogram normal fibroblasts into cancer-associated fibroblasts in ovarian cancer

    Cancer Discov

    (2012)
  • B.F. Ning et al.

    Hepatocyte nuclear factor 4 alpha suppresses the development of hepatocellular carcinoma

    Cancer Res

    (2010)
  • J. Wang et al.

    FOXA2 suppresses the metastasis of hepatocellular carcinoma partially through matrix metalloproteinase-9 inhibition

    Carcinogenesis

    (2014)
  • M. Ota et al.

    Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling

    Development

    (2008)
  • W.P. Xu et al.

    Perturbation of MicroRNA-370/Lin-28 homolog A/nuclear factor kappa B regulatory circuit contributes to the development of hepatocellular carcinoma

    Hepatology

    (2013)
  • H. Sasaki et al.

    A binding site for Gli proteins is essential for HNF-3beta floor plate enhancer activity in transgenics and can respond to Shh in vitro

    Development

    (1997)
  • R. Kalluri et al.

    The basics of epithelial-mesenchymal transition

    J. Clin. Invest

    (2009)
  • F. Marra et al.

    Roles for chemokines in liver disease

    Gastroenterology

    (2014)
  • R. Galuppo et al.

    Molecular therapies in hepatocellular carcinoma: what can we target

    Dig. Dis. Sci

    (2014)
  • Cited by (171)

    • Cancer-Associated Fibroblasts in Hepatocellular Carcinoma and Cholangiocarcinoma

      2023, Cellular and Molecular Gastroenterology and Hepatology
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text