SPOCK1 is up-regulated and promotes tumor growth via the PI3K/AKT signaling pathway in colorectal cancer

https://doi.org/10.1016/j.bbrc.2016.11.126Get rights and content

Highlights

  • SPOCK1 expression is significantly up-regulated in CRC tissues and associated with tumor size and TMN stage.

  • Down-regulation of SPOCK1 suppresses CRC cell proliferation both in vitro and in vivo.

  • SPOCK1 stimulates the PI3K/AKT signaling pathway to inhibit cell apoptosis and promote tumor growth.

Abstract

SPOCK1 encodes a Ca2+-binding matricellular glycoprotein which plays an oncogenic role in cancer cells. However, the role of SPOCK1 in the pathogenesis of colorectal cancer (CRC) has not been determined. Here, SPOCK1 was found higher expressed in CRC tissues than that of adjacent normal tissues. Furthermore, up-regulated expression of SPOCK1 in CRC patients was associated with tumor size and TNM stage. In addition, we observed that the depletion of SPOCK1 inhibited proliferation in vitro and tumorigenicity in vivo and promoted apoptosis in cell culture. Our data suggest that inactivation of PI3K/Akt signaling pathway was involved in down-regulation of SPOCK1-mediated suppression of tumor cell proliferation. These results suggest that SPOCK1 expression is correlated with malignant features of CRC and may serve as potential therapeutic and preventive strategies for CRC.

Introduction

Colorectal cancer(CRC) is one of the most common malignant cancers, the third leading cause of cancer-related death in men(746,000 cases, 10% of all cancers), the second leading cause of cancer-related death in women (614,000 cases, 9.2% of all cancers), and approximately 600,000 people died of CRC annually worldwide [1]. The increasing prevalence of virulence factors for CRC such as dietary factors, obesity, environmental exposures, genetic susceptibility and smoking are gradually being determined [2], [3]. Despite advances in early detection, surgical techniques, chemotherapy, radiotherapy, molecular targeted therapy have increased the prognosis of CRC patient, the 5-year survival rate is less than 40% in less developed countries, including China [4]. Molecular signaling and proliferation of tumors have also been shown to be closely associated with CRC, but the molecular mechanisms underlying CRC carcinogenesis are still under investigation. Thus, novel molecular biomarkers to better understand CRC proliferation as well as cancer therapeutic targets should be explored.

The SPARC/osteonectin, cwcv, and kazal-like domain proteoglycan 1 (SPOCK1) was first identified in human testicular seminal plasma, which encodes a Ca2+-binding matricellular glycoprotein that belongs to the SPARC family [5], [6]. SPOCK1, as well as other members of this family, which shares a similar structure that consists of N-terminus, follistatin-like domain, and C-terminus, and plays crucial roles in cell proliferation, cell–cell adhesion and migration [7]. Accumulating evidence has confirmed that amplification of SPOCK1, as well as mRNA and protein overexpression, involves in the regulation of cell cycle control, apoptosis, and metastasis, which has been frequently shown in various human malignancies, such as prostate, gallbladder, lung carcinomas [8], [9], [10]. However, to the best of our knowledge, information on the relationship between SPOCK1 and CRC, including the molecular mechanisms and clinical potentials underlying the participation of SPOCK1 in these malignant features, are limited.

In the current work, we first revealed that expression of SPOCK1 was up-regulated in CRC patients. Second, we show that SPOCK1 depletion effectively inhibits cell proliferation in vitro and significantly suppresses tumor growth in vivo. In addition, cell proliferation was suppressed by inducing cell cycle arrest at the G0/G1 phase in SPOCK1-knockdown cells. Last, we demonstrate that the PI3K/AKT pathway is involved in the oncogenic function of SPOCK1-mediated cell survival. This research provided new understanding into the pathogenesis of CRC which will constitute a potential biomarkers and aid novel therapeutic strategies.

Section snippets

CRC patient specimens

To examine the expression of SPOCK1 in human CRC, the paired tumorous and adjacent non-tumorous CRC specimens which were obtained from 84 CRC patients who underwent radical resection (including lymph-adenectomy) without prior chemotherapy or radiotherapy at the Department of General Surgery, Affiliated Hospital of Nantong University. Resected CRC tumor samples and matched adjacent non-tumor tissues were collected and immediately stored in liquid nitrogen. All tissue samples were confirmed for

SPOCK1 expression in CRC tissues and cells

To explore the role of SPOCK1 in the progression of CRC, we first detected SPOCK1 mRNA expression of 84 pairs of CRC samples by qRT-PCR. It was shown that SPOCK1 expression was significantly higher in the CRC tissues compared with matched non-cancerous tissues (Fig. 1A). SPOCK1 mRNA expression levels in five CRC cancer cell lines and normal human colon epithelial cells were also evaluated by using qRT-PCR (Fig. 1B). SPOCK1 mRNA expression was increased in all CRC-derived cell lines (SW480, HT29

Discussion

Although significant progress has been made for the prognosis of CRC in the early stage, the majority of CRC patients are usually diagnosed at an advanced stage which leads to a poor prognosis [14]. The relapse rate of patients with CRC is about 30–50% [15]. The mechanisms underlying CRC development, progression and recurrence is complex. Thus, identification of preventive strategies and therapeutic targets is urgent for CRC patients. In this study, we identified a new potential biomarker,

Competing interests

The authors declared that they have no competing interests.

Acknowledgment

This work was funded by the Scientific Innovation Foundation of Nantong (HS2014043).

References (31)

  • P.M. Alliel et al.

    Testican, a multidomain testicular proteoglycan resembling modulators of cell social behaviour

    Eur. J. Biochem.

    (1993)
  • Y. Li et al.

    SPOCK1 is regulated by CHD1L and blocks apoptosis and promotes HCC cell invasiveness and metastasis in mice

    Gastroenterology

    (2013)
  • D. Ahel et al.

    Poly(ADP-ribose)-dependent regulation of DNA repair by the chromatin remodeling enzyme ALC1

    Science

    (2009)
  • Q. Chen et al.

    SPOCK1 promotes tumor growth and metastasis in human prostate cancer

    Drug Des. Devel Ther.

    (2016)
  • Y.J. Shu et al.

    SPOCK1 as a potential cancer prognostic marker promotes the proliferation and metastasis of gallbladder cancer cells by activating the PI3K/AKT pathway

    Mol. Cancer

    (2015)
  • Cited by (0)

    1

    These authors contributed equally to this article.

    View full text