Molecular and Cellular Pharmacology
Mechanistic role of p38 MAPK in gastric cancer dissemination in a rodent model peritoneal metastasis

https://doi.org/10.1016/j.ejphar.2011.11.015Get rights and content

Abstract

Peritoneal dissemination is a highly frequent complication of poorly differentiated gastric cancers for which no effective therapies are available. Constitutive activation of mitogen-activated protein kinases (MAPKs) signaling cascades is recognized as a causative factor in the malignant transformation of several carcinoma cell types. In the present study we provide evidence that p38 MAPK inhibition protects against gastric cancer cells dissemination in a mouse model of peritoneal carcinomatosis. Administering mice with ML3403 and SB203580, potent and selective p38 MAPK inhibitors, attenuate the formation of neoplastic foci induced by intraperitoneal inoculation of gastric cancer cells. By gene array analysis we found that such a protective effect correlates with a robust downregulation in the expression of CXC chemokine receptor-4, Fms-related tyrosine kinase 4 (FLT4), the non-receptor spleen tyrosine kinase (SYK) and the collagen α2(IV) (COL4A2) in neoplasic foci. Inhibition of p38 MAPK in vivo increased the sensitivity of tumor cells to cisplatin and associated with a robust downregulation in the expression of the multidrug resistance (MDR)-1, a well defined marker of resistance to chemotherapy. In summary, p38 MAPK inhibition by a small molecule is beneficial in preventing the peritoneal dissemination of poorly differentiated gastric cancer cells by acting at multiple check-points in the process of attachment and diffusion of tumor cells in the peritoneum.

Introduction

MAPKs are serine/threonine kinases activated in response to a variety of external signals (Cuadrado and Nebreda, 2010, Olson and Hallahan, 2004, Wagner and Nebreda, 2009). Three major subclasses of MAPKs, namely, extracellular signal-regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p38 have been identified. Various receptor tyrosine kinases, cytokine receptors, G proteins and oncogene products can activate MAPKs. Thus, MAPKs are proposed to be a critical integrator of various signaling transduction systems and involved in various cellular processes including cell proliferation, differentiation, apoptosis, and transformation (Cuadrado and Nebreda, 2010, Olson and Hallahan, 2004, Wagner and Nebreda, 2009).

Constitutive activation of these signaling cascades has been reported as a major causative factor in the malignant transformation of cancer cell lines (Leav et al., 1996, Salh et al., 1999) and is implicated in the carcinogenesis and metastatic potential of human cancers (Huang et al., 2000, Ito et al., 1998, Miki et al., 1999). Thus, there is robust evidence that tumor cell invasion associates with activation of p38 MAPK signaling pathway in cancers of the head and neck, breast, colon (Junttila et al., 2007) and melanoma (Boukerche et al., 2005, Boukerche et al., 2010).

Human scirrhous gastric carcinoma is a diffusely infiltrating type of poorly differentiated gastric carcinoma which, histopathologically, do not form glands, but growth by diffuse infiltration of a broad region of the gastric wall, resulting in fibrous-like thickening of the gastric wall (Dicken et al., 2005) and peritoneal dissemination or distant metastasis to lymph nodes has already occurred in many cases after clinical diagnosis. There are no standard treatments for peritoneal dissemination (Dicken et al., 2005, Yonemura et al., 2007). Peritoneal dissemination occurs frequently even after radical surgery, and is a major factor determining the poor prognosis of patients with scirrhous gastric carcinoma (Dicken et al., 2005, Yonemura et al., 2007). Further systemic chemotherapy tends to have little effect on peritoneal dissemination, because the peritoneal–blood barrier hinders drug distribution throughout the peritoneal cavity (Yonemura et al., 2007). To date, however, the mechanism of peritoneal dissemination of gastric cancer has not yet been fully elucidated. High levels of phospho-p38 MAPK are detected in poorly differentiated gastric cancers in comparison to differentiated cancers (Atsumi et al., 2007, Yokozaki, 2007). It has been suggested that the p38 MAPK signaling may be implicated in the disarrangement of cytoskeletal F-actin filaments and transcriptional suppression of E-cadherin in poorly differentiated gastric cancers. These effects may lead to an irregular cell shape and loss of cell–cell adhesion.

Here, we report that the p38 MAPK pathway plays a mechanistic role in peritoneal dissemination of human gastric cancer cells. By demonstrating that pharmacological inhibition of p38 MAPK prevents metastatic dissemination of gastric cancer cells and have an additive effect in the sensitivity to the chemotherapy, this study suggests that small molecules p38 MAPK inhibitors have anti-cancer potential.

Section snippets

Cell line and treatment

Human gastric cancer cell lines (Yokozaki, 2007), MKN74 and MKN45, were purchased from the Japanese Collection of Research Bioresources, Human Science Research Resources Bank (Osaka, Japan). Gastric cell lines, were maintained in RPMI medium with 10% fetal bovine serum (FBS) and penicillin/streptomycin at 37 C in a humidified atmosphere of 5% CO2 in air. Cells were regularly passaged to maintain exponential growth. For in vitro study ML3403 and SB203580 (both provided by Stefan Laufer,

Pospho-p38 MAPK levels and peritoneal gastric cancer dissemination

It has been (Atsumi et al., 2007) suggested that p38 MAPK is expressed in poorly differentiated gastric carcinomas and expression correlates with the potential for peritoneal dissemination. As illustrated in Fig. 1A, and in agreement with previously published data (Atsumi et al., 2007) poorly differentiated, diffuse-type, gastric carcinomas stained positively with antibodies detecting the total and the phosphorylated form of p38 MAPK. The p38 MAPK expression was mainly found in the neoplastic

Discussion

In the present study we have shown that p38 MAPK inhibition prevents peritoneal dissemination of non-differentiated gastric cancer cells in a mouse model of peritoneal carcinomatosis. In addition, we have provided evidence that p38 inhibition increases the chemosensitivity to cisplatin and might have utility in the treatment of chemoresistant gastric tumors.

The p38 MAPK pathway plays a key role during the early stages of invasion and metastasis of carcinoma cells (Rosivatz et al., 2002,

Authors' contributions

LG and AM designed the study, contributed to the experimental work and wrote the manuscript. CS and EC contributed animal data. SC contributed IHC staining data. BR contributed microarray analysis. GP contributed RT-PCR and Western blot data. SL provided chemicals used in these studies. MB, AD and SF designed the study, contributed to data interpretation and wrote the manuscript. All authors have read and approved the final manuscript.

The following are the supplementary materials related to

References (37)

  • H. Boukerche et al.

    Src kinase activation is mandatory for MDA-9/syntenin-mediated activation of nuclear factor-Kb

    Oncogene

    (2010)
  • T. Boulikas et al.

    Cisplatin and platinum drugs at the molecular level

    Oncol. Rep.

    (2003)
  • A. Cuadrado et al.

    Mechanisms and functions of p38 MAPK signalling

    Biochem. J.

    (2010)
  • B.J. Dicken et al.

    Gastric adenocarcinoma: review and considerations for future directions

    Ann. Surg.

    (2005)
  • X. Guo et al.

    Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells

    BMC Cancer

    (2008)
  • B. Ingold et al.

    Vascular CXCR4 expression — a novel antiangiogenic target in gastric cancer?

    PLoS One

    (2010)
  • Y. Ito et al.

    Activation of mitogen-activated protein kinases/extracellular signal-regulated kinases in human hepatocellular carcinoma

    Hepatology

    (1998)
  • M.R. Junttila et al.

    p38a and p38δ mitogen-activated protein kinase isoforms regulate invasion and growth of head and neck squamous carcinoma cells

    Oncogene

    (2007)
  • Cited by (25)

    • MAPK14 (p38α) inhibition effects against metastatic gastric cancer cells: A potential biomarker and pharmacological target

      2020, Toxicology in Vitro
      Citation Excerpt :

      The activation of MAPKs can be performed by tyrosine kinases receptors, cytokine receptors, G proteins, and oncogene products. Because of their role in diverse cellular processes, their dysregulation often leads to disease (Graziosi et al., 2012). The MAPK14/p38 family has been reported to be affected by environmental stress.

    • Cancer and Microenvironment Plasticity: Double-Edged Swords in Metastasis

      2019, Trends in Pharmacological Sciences
      Citation Excerpt :

      In line with this, other EMT-regulating factors, such as Y-box binding protein 1 (YB-1) [16,24] and grainyhead-like transcription factor 2 (GRHL2) [17,18], could also play dichotomous roles in tumor growth and metastasis. In addition, some other molecules such as homeobox C9 (HOXC9) [25], p38 mitogen-activated protein kinase (MAPK) [26,27], and ephrin type-B receptor 2 (EphB2) [28,29] (summarized in Table 1), although not implicated in EMT manipulation, could also regulate cancer cell proliferation and migration oppositely. These opposite functions of the above modulators usually result from their ability to regulate multiple downstream pathways that are relevant to proliferation and metastasis.

    • Increased serum chemerin level promotes cellular invasiveness in gastric cancer: A clinical and experimental study

      2014, Peptides
      Citation Excerpt :

      p38 MAPK also plays a key role in the pathogenesis of gastric cancer [3]. p38 activation has been shown to accelerate peritoneal dissemination by enhancing attachment and diffusion of gastric cancer cells in the peritoneum [17]. The association between chemerin and p38 activation has been demonstrated in primary human granulosa cells and endothelial cells [26,36].

    • Chemerin and cancer

      2019, International Journal of Molecular Sciences
    View all citing articles on Scopus
    1

    Contributed equally to this work.

    View full text