Open Access

The role of human peritoneal mesothelial cells in the fibrosis and progression of gastric cancer

  • Authors:
    • Tomoya Tsukada
    • Sachio Fushida
    • Shinichi Harada
    • Yasumichi  Yagi
    • Jun  Kinoshita
    • Katsunobu Oyama
    • Hidehiro Tajima
    • Hideto  Fujita
    • Itasu Ninomiya
    • Takashi Fujimura
    • Tetsuo Ohta
  • View Affiliations

  • Published online on: May 18, 2012     https://doi.org/10.3892/ijo.2012.1490
  • Pages: 476-482
  • Copyright: © Tsukada et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Peritoneal dissemination is the most frequent metastatic pattern of scirrhous gastric cancer. However, despite extensive research effort, disease outcomes have not improved sufficiently. Tumor progression and metastasis result from interactions between cancer and various cells in the stroma, including endothelial cells, immune cells and fibroblasts. Fibroblasts have been particularly well studied; they are known to change into carcinoma-associated fibroblasts (CAFs) and produce transforming growth factor β (TGF-β), which mediates cancer-stroma interactions. Here, we investigated whether TGF-β derived from cancer cells in the peritoneal microenvironment activates human peritoneal mesothelial cells (HPMCs), leading to the progression and fibrosis of gastric cancer. We found that activated HPMCs (a-HPMCs) took on a spindle shape formation, decreased the expression of E-cadherin and increased that of α-SMA. Furthermore, a-HPMCs became more invasive and upregulated proliferation of human gastric cancer-derived MKN45 cells following direct cell-cell contact. Notably, MKN45 cells co-cultured with a-HPMCs also acquired anchorage-independent cell growth and decreased expression of E-cadherin in vitro. To measure the effects of the co-culture in vivo, we developed a mouse xenograft model into which different culture products were subcutaneously injected. The largest tumors were observed in mice that had been given MKN45 cells co-cultured with a-HPMCs. Furthermore, these tumors contained HPMC-derived fibrous tissue. Thus, the epithelial-mesenchymal transition (EMT) of HPMCs appears to drive peritoneal dissemination and tumor fibrosis.

Introduction

The diffusely infiltrating human scirrhous gastric carcinoma is characterized by cancer cell infiltration and proliferation accompanied by extensive stromal fibrosis. The most frequent metastatic pattern of scirrhous gastric carcinoma is peritoneal dissemination, for which anticancer therapies have been developed. However, despite the proven benefits of these treatments (1,2), outcomes of this carcinoma require further improvements.

Although cancer research has traditionally focused on malignant cancer cells, recent developments have shown that tumor progression is the product of interactions between cancer cells and various cells in the stroma, such as endothelial cells, immune cells, and fibroblasts (36). Fibroblasts have been particularly well-studied (7); for instance, the presence of orthotopic fibroblasts has been shown to contribute to cell growth and extensive stromal fibrosis at the primary cancer site (8). Furthermore, it is known that growth factors derived from orthotopic fibroblasts are not adequate for enhancing gastric cancer invasiveness; a direct interaction between cancer cells and orthotopic fibroblasts is important for acceleration of cancer cell invasion and progression (9). At the peritoneal dissemination site, cancer cells usually generate a supportive microenvironment by producing stroma-modulating growth factors (fibroblast growth factor [FGF] family, platelet-derived growth factor [PDGF], epidermal growth factor [EGF] ligands, vascular endothelial growth factor [VEGF] family, interleukins, and transforming growth factor-β1 [TGF-β1]) (10,11). These factors activate fibroblasts and facilitate additional secretion of growth factors and proteases. As a result, scirrhous gastric cancer cells stimulate the proliferation of peritoneal fibroblasts and induce peritoneal fibrosis.

Fibroblasts within the cancer stroma are known as carcinoma-associated fibroblasts (CAF), and acquire a modified phenotype (12,13). Cancer cells are modified by CAFs and induce epithelial-mesenchymal transition (EMT)-like changes characterized by repression of E-cadherin expression and enhancement of α-SMA expression. Generally, cell transformation progresses gradually, rather than drastically in the EMT process. However, under these cell-cell interactions, each cell is facilitated by a characteristic change into irreversible transformation (14).

Fibroblasts are the principal effectors mediating fibrosis. In the tumor microenvironment, activated fibroblasts arise from several sources, such as resident fibroblasts or myofibroblasts (15), bone marrow-derived cells or fibrocytes (3,16), and both endothelial- and epithelial-mesenchymal transitions (EndMT and EMT, respectively) (5).

In scirrhous gastric cancer, fibrous tissue volume and composition are regulated by the response of fibroblasts to the growth factors that are released by cancer cells (17,18). Increased expression of TGF-β1, in particular, has been demonstrated in fibrotic tissue and regions of increased extra-cellular matrix deposition in various organs (19,20). TGF-β1 is also known as the crucial inducer of EMTs, which play an important role in the development and malignancy of various human carcinomas (21). Loss of epithelial character is typically observed late in the progression of human carcinomas, and correlates with their growth, local invasion, angiogenesis, and metastasis (22).

Human peritoneal mesothelial cells (HPMCs), which are classified as epithelium in the broad sense of the term, serve as a protective anatomical barrier and play a key role in the immunological response to infection, wound healing, and the dissemination of tumor cells such as those responsible for ovarian cancers (23). HPMCs also produce EGF, FGF and VEGF, and their expression is significantly upregulated in the peritoneal cavity following surgical intervention, when free cancer cells may be spilled during the course of the resection (24). In cancerous environments, HPMCs frequently undergo a change in morphology, taking on a fibroblastic shape rather than their usual epithelial-like formation - a change that, along with fibrosis of the peritoneum, was recently linked to TGF-β1 activity (25,26). These physical alterations expose submesothelial connective tissue and facilitate adhesion of cancer cells (27). Activated HPMCs also acquire migratory potential (25), but their proliferation potential and their possible interactions with cancer cells remains unknown.

In this study, we investigated whether activated HPMCs (a-HPMC) induced by TGF-β1 stimulation contribute to gastric cancer cell infiltration, proliferation, and fibrosis. We documented the behavior of HPMC, a-HPMC, and gastric cancer cells in the presence or absence of direct co-culture with HPMCs. We also used a mouse xenograft model to study the interactions of gastric cancer cells and HPMCs or a-HPMCs. We found that HPMCs exposed to TGF-β1 acquire a fibroblast-like formation and the ability to invade. Furthermore, presence of a-HPMCs contributes to cancer cell growth and fibrosis under conditions of direct cell contact in the tumor microenvironment.

Materials and methods

Cell lines and cell culture

HPMCs were isolated from surgical specimens of human omentum as previously described (28). Omental specimens were obtained, with informed consent, from patients undergoing elective abdominal surgery. Tissue samples were collected in ice-cold phosphate buffered saline (PBS) to minimize cell degeneration. Samples were immediately washed extensively in PBS to remove contaminating red blood cells and incubated with pre-warmed PBS containing 0.125% trypsin/EDTA (Gibco, USA) for 30 min at 37°C. The suspension was passed through a 100-μm-pore nylon mesh (Becton Dickinson, Japan) to remove undigested fragments, then centrifuged at 1,500 rpm for 5 min. The collected cells were cultured in RPMI 1640 medium (Gibco) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Nichirei Bioscience Inc., Japan), 100 IU/ml penicillin, 100 mg/ml streptomycin (Gibco), and 2 mM glutamine (Nissui Pharmaceutical Co., Ltd., Japan). Cells were seeded in a gelatin-coated 75-cm2 dish flask (BD Falcon, USA) and cultured in 10 ml of medium at 37°C in a humidified atmosphere of 5% CO2 in air.

Human gastric cancer cell lines (MKN45) were obtained from the American Type Culture Collection (Rockville, USA). The culture medium for MKN45 cells was RPMI-1640 (Gibco) with the same additives as mentioned above. MKN45 cells were grown to confluence and harvested by trypsinization with 0.25% trypsin/EDTA. The confluent HPMCs were trypsinized with 0.125% trypsin/EDTA prior to use. HPMCs were then transferred to serum-free medium for 24 h, after which they were continuously exposed to 5 ng/ml of recombinant human TGF-β1 (Sigma-Aldrich, Inc., USA) for 48 h. Finally, they were transferred to RPMI-1640 containing 10% FBS, which caused the cells to undergo a shift in morphology, allowing us to identify the resulting cells as activated HPMCs (a-HPMCs). HPMCs were used at passage 1–3 in all experiments.

Invasion assay

The effects of TGF-β1 treatment on cell invasion were determined using a BD BioCoat Matrigel Invasion Chamber for 24-well plates (BD Bioscience, USA), according to the manufacturer’s instructions. First, Matrigels were rehydrated using 750 μl of serum-free medium, after which we added 750 μl of fresh medium containing 10% FBS to the lower chamber. Next, 0.5 ml of HPMC and a-HPMC cells (1×105 cells/ml) in serum-free media were seeded into the upper chamber of the system (both the control membrane and Matrigel membrane were seeded with cells). After 24 h of incubation, the cells in the upper chamber were removed and the cells that had invaded through the Matrigel membrane were stained by hematoxylin and fixed in 100% methanol. Membranes were removed from inserts and mounted on slides. Invading cells were counted using a microscope with a 10× objective in several fields of triplicate membranes. Data are expressed as the percentage of invasion through the Matrigel membrane relative to the migration through the control membrane.

Cell proliferation assay

MKN45 cells, seeded at a density of 1×105 cells per well in 6-well plates, were incubated alone (control) or in the presence of a direct co-culture with the same number of HPMCs or a-HPMCs. A 1-μm pore Boyden Chamber (BD Falcon) was used for indirect incubation. Cells were counted on Days 1, 3, and 5 post-seeding. The magnetic-activated cell sorting (MACS, Miltenyi Biotec, Germany) method was used to separate MKN45 cells from HPMCs and a-HPMCs. Following trypsinization, microbead-labeled anti-human CD326 antibody (Miltenyi Biotec) was used to label cells. Data were averaged across each experiment (for example, ‘with co-culture’ and ‘without co-culture’), for which sample points were assayed in triplicate and results were counted twice.

Transformation assay

Soft agar assays were performed in 6-well plates. The assay medium comprised equal volumes of 2-fold concentration RPMI-1640 containing 20% FBS and 1.44% agar solution (final concentration = 0.72%). Individual wells were coated with 1.5 ml of base medium.

The cells (HPMC, a-HPMC, MKN45, MKN45 co-cultured with HPMC, MKN45 co-cultured with a-HPMC) were harvested with trypsin, resuspended in RPMI-1640 containing 10% FBS, and mixed with an equal volume of the aforementioned assay medium (final concentration = 0.36%). Co-culture cells were incubated for 5 days and isolated by MACS. In all soft agar cultures, cells were seeded on solidified base layers in a 2-ml volume at a density of 1×104 cells/ml. These cultures were incubated for 14 days and counted using a microscope with a 4× objective in several fields. Colonies containing >5 cells were scored as positive. All examinations were performed in triplicate.

Western blotting

Cells were lysed in RIPA buffer (50 mmol/l Tris-HCl (pH 8.0), 150 mmol/l sodium chloride, 0.5 w/v% sodium deoxycholate, 0.1 w/v% sodium dodecyl sulfate, 1.0 w/v% NP-40 substitute) (Wako, Japan) containing 1% protease inhibitor cocktail (Sigma-Aldrich Inc.). The protein concentration of each sample was measured using a BCA protein assay kit (Pierce Biotechnology, USA). Whole-cell lysates were prepared in denaturing SDS sample buffer and subjected to SDS-PAGE (ATTO Co. Ltd., Japan). The immunoblots were visualized using an ECL Plus kit (GE Healthcare UK Ltd.). We employed the following primary antibodies: E-cadherin (H-108, rabbit polyclonal IgG, diluted 1:1,000; Santa Cruz Biochemistry, USA), α-SMA (1A4, mouse monoclonal IgG, diluted 1:5,000; DakoCytomation, Denmark), and β-actin (AC-15, mouse monoclonal IgG, diluted 1:10,000; Sigma).

Mouse xenograft model

All animal experiments were performed according to Kanazawa University’s standard guidelines. We used BALB/c nu/nu mice (female, 4–6 weeks old; Charles River Laboratories Inc. Japan) as a xenograft model in which we investigated the effects of co-culturing MKN45 with HPMCs and a-HPMCs. HPMCs and a-HPMCs were first stained with a red fluorescent dye PKH26 cell linker kit (Sigma) according to the manufacturer’s instructions; the concentration of PKH26 during incubation was 4 μM. Next, MKN45 cells were co-cultured with an equivalent number of HPMCs or a-HPMCs for 5 days. A total of 5×106 cells in 100 μl of RPMI-1640 were then subcutaneously injected into the dorsal side of each mouse (n=27). The mice were divided into the following experimental groups: a control group (MKN45 s.c., n=9), MKN45 co-cultured with HPMC group (n=9), and MKN45 co-cultured with a-HPMC group (n=9). Xenograft tumors were measured every other day for 10 days. Tumor volume was estimated using the equation v=(ab2)/2, where v is volume, a is the length of the major axis, and b is the length of the minor axis. At the end of the experiment, tumor specimens were collected for immunohistochemical examination.

Histological and immunohistochemical examination

Tumor specimens obtained from subcutaneous tumors were shock frozen in liquid nitrogen for fluorescence microscopy. Specimens were cryosectioned and mounted on a glass slide, air dried, and immediately analyzed by fluorescence microscopy using a standard filter setup for visualization of PKH26. Tumor specimens were then fixed in 10% neutral buffered formalin and embedded in paraffin. Sections were stained with hematoxylin and eosin (H&E) and Azan stain, and immunostained with E-cadherin antibody (H-108, rabbit polyclonal IgG, diluted 1:100; Santa Cruz Biochemistry) and α-SMA (1A4, mouse monoclonal IgG, diluted 1:100; DakoCytomation) at 4°C overnight. The sections were treated with EnVision reagent (Dako Co., Japan) for visualization.

Statistical analysis

We investigated differences among the data sets by one-way analysis of variance (ANOVA) or two-sided Student’s t-tests using the computer software package SPSS 10.0 (SPSS Inc., USA). P<0.05 indicated a statistically significant difference.

Results

Effect of TGF-β1 on the invasiveness and anchorage-independent growth of human peritoneal mesothelial cells in vitro

Morphological changes in cultured HPMCs were observed 48 h after the addition of TGF-β1. HPMCs cultured without TGF-β1 had a cobblestone-like appearance (Fig. 1A, left). By contrast, HPMCs treated with TGF-β1 displayed a spindle fibroblastic pattern morphology (Fig. 1A, right). These changes are typical of cells with a mesenchymal phenotype. TGF-β1 exposure increased the invasiveness of HPMCs (P<0.001; Fig. 1B). As expected, given the morphological changes, western blot analysis showed a decrease in E-cadherin expression and an increase in α-SMA expression (Fig. 2A).

HPMCs were not able to grow in soft agar gel, regardless of whether or not TGF-β1 was present (Fig. 3A and B). Thus, treatment with TGF-β1 was responsible for the acquired abilities of mobility and invasiveness through the basal membrane. However, HPMCs did not independently acquire anchorage-independent growth.

Effect of a-HPMC co-culture on the proliferation of gastric cancer cells

MKN45 cell count was elevated after co-culturing with a-HPMCs (P<0.001; Fig. 2B). However, when MKN45 and a-HPMCs were separately cultured using Boyden Chamber inserts, no significant increase in MKN45 cells was detected (Fig. 2B). In addition, MKN45 cells co-cultured with a-HPMCs were able to form larger and greater colonies in soft agar gel (Fig. 3A and B).

No morphological changes of MKN-45 cells during this assay were observed (data not shown). However, our results showed that cell-cell contact with HPMCs attenuates intra-cellular expression of E-cadherin (Fig. 2A) in MKN45 cells. Elevation of α-SMA expression was also observed in MKN45 cells co-cultured with a-HPMCs. This result indicates that the MACS method was not able to eliminate cell contamination.

Effects of a-HPMCs in subcutaneous xenograft models

The time course of subcutaneous relative tumor volume is shown in Fig. 4A. At 10 days post-inoculation, the mean relative volume of tumors created with MKN45 cells co-cultured with a-HPMCs was significantly larger than that of the other groups (P<0.001). However, we did not observe any morphological differences in the subcutaneous tumors (for example, scirrhous or medullary types) (Fig. 4B). The area of fibrosis was significantly larger in tumors created from MKN45 cells co-cultured with a-HPMCs than in tumors from the other groups (Fig. 5B). We confirmed implantation of the subcutaneous tumors and HPMCs labeled by PKH26 cell linker kit (Fig. 5C); in tumors from MKN45 cells co-cultured with a-HPMCs, there were higher numbers of implanted cells and higher levels of fibrosis.

Expression of α-SMA in the subcutaneous tumors was highest in animals implanted with MKN45 cells co-cultured with a-HPMCs, which corresponds to the notable increase in fibrous tissue (Fig. 5D). Expression of E-cadherin was lower in MKN45 cells co-cultured with both HPMCs and a-HPMCs than in MKN45 cells cultured alone. This suggests that HPMCs were responsible for the reduction in E-cadherin expression (Fig. 5E).

Discussion

We have shown that tumor fibrosis is produced not only by orthotopic fibroblasts, but also by HPMCs undergoing mesenchymal transition; furthermore, we have demonstrated that direct physical contact with cancer cells drives the process of fibrosis (Fig. 6). HPMCs stimulated by TGF-β1 undergo morphological changes (Fig. 1A) and also acquire the ability to invade the basal membrane in vitro (Fig. 1B). We found that direct contact between gastric cancer cells and a-HPMCs contributes to the growth and transformation of gastric cancer cells (Figs. 2 and 3). As a result, activity of a-HPMCs on the basal membrane causes highly fibrotic changes in vivo (Figs. 4 and 5).

Compelling evidence suggests that morphological changes in HPMCs are caused by TGF-β1 derived from gastric cancer cells and host fibroblasts (29,30); expression of this protein causes the exposure of submesothelial connective tissue and adhesion of cancer cells. In our current mouse xenograft model, a-HPMCs implanted in greater quantities, and induced more fibrosis than HPMCs (Figs. 5A and B). a-HPMCs acquire invasiveness in vitro and contribute to cancer cell proliferation as CAFs do (Figs. 1B and 2). CAFs are derived from various cells, including local fibroblasts, bone marrow fibrocytes, and endothelial cells (3,5,16). Our study is the first to suggest that a-HPMCs are also a possible source of CAFs.

We found that direct contact with a-HPMCs led to anchorage-independent growth (Fig. 3) and repression of E-cadherin expression (Figs. 2 and 5) in gastric cancer cells. This indicates that a-HPMCs contribute to the EMT-like change observed in gastric cancer cells following cell-cell contact. However, cancer cells co-cultured with normal HPMCs also have reduced E-cadherin expression (Fig. 2). According to recent reports, CAFs are implicated in cancer by producing high quantities of stromal-derived factor-1α (SDF-1α), also known as chemokine CXCL12 (31); SDF-1α expression is higher in mesothelial cells than in other organs (32). Although we hypothesized that differences in SDF-1α expression may be responsible for the differential activity of HPMCs and a-HPMCs observed in our study, we found no evidence to support this (data not shown). Thus, future research should attempt to elucidate the mechanism behind the different effects of HPMCs and a-HPMCs on gastric cancer cells.

In conclusion, we have shown that HPMCs activated by TGF-β1 signaling acquire their own invasiveness and promote fibrosis in a mouse xenograft model. In addition, direct contact with a-HPMCs contributes to EMT-like changes in gastric cancer cells and promotes tumor growth. Our results strongly suggest that HPMCs are one of the origins of CAFs; therefore, understanding the mechanism of EMT in HPMCs is necessary for developing a molecular approach to combat peritoneal dissemination of gastric cancer.

Acknowledgments

We are grateful to members of the Department of Gastroenterologic Surgery of Kanazawa University for their helpful suggestions. We thank Dr Tomohiko Wakayama and Professor Shoichi Iseki for providing technical support during fluorescence microscopy.

References

1 

Fushida S, Kinoshita J, Yagi Y, et al: Dual anti-cancer effects of weekly intraperitoneal docetaxel in treatment of advanced gastric cancer patients with peritoneal carcinomatosis: A feasibility and pharmacokinetic study. Oncol Rep. 19:1305–1310. 2008.

2 

Yonemura Y, Elnemr A, Endou Y, et al: Multidisciplinary therapy for treatment of patients with peritoneal carcinomatosis from gastric cancer. World J Gastrointest Oncol. 2:85–97. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Kalluri R and Zeisberg M: Fibroblasts in cancer. Nat Rev Cancer. 6:392–401. 2006. View Article : Google Scholar

4 

Tlsty TD: Stromal cells can contribute oncogenic signals. Semin Cancer Biol. 11:97–104. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Zeisberg EM, Potenta S, Xie L, Zeisberg M and Kalluri R: Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 67:10123–10128. 2007.PubMed/NCBI

6 

Pietras K and Ostman A: Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 316:1324–1331. 2010. View Article : Google Scholar : PubMed/NCBI

7 

De Wever O and Mareel M: Role of tissue stroma in cancer cell invasion. J Pathol. 200:429–447. 2003.PubMed/NCBI

8 

Yashiro M, Chung YS and Sowa M: Role of orthotopic fibroblasts in the development of scirrhous gastric carcinoma. Jpn J Cancer Res. 85:883–886. 1994. View Article : Google Scholar : PubMed/NCBI

9 

Semba S, Kodama Y, Ohnuma K, et al: Direct cancer-stromal interaction increases fibroblast proliferation and enhances invasive properties of scirrhous-type gastric carcinoma cells. Br J Cancer. 101:1365–1373. 2009. View Article : Google Scholar

10 

Nakazawa K, Yashiro M and Hirakawa K: Keratinocyte growth factor produced by gastric fibroblasts specifically stimulates proliferation of cancer cells from scirrhous gastric carcinoma. Cancer Res. 63:8848–8852. 2003.

11 

Elenbaas B and Weinberg RA: Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp Cell Res. 264:169–184. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Durning P, Schor SL and Sellwood RA: Fibroblasts from patients with breast cancer show abnormal migratory behaviour in vitro. Lancet. 2:890–892. 1984. View Article : Google Scholar : PubMed/NCBI

13 

Schor SL, Schor AM, Grey AM and Rushton G: Foetal and cancer patient fibroblasts produce an autocrine migration-stimulating factor not made by normal adult cells. J Cell Sci. 90:391–399. 1988.PubMed/NCBI

14 

Grünert S, Jechlinger M and Beug H: Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol. 4:657–665. 2003.PubMed/NCBI

15 

Brenmoehl J, Miller SN, Hofmann C, et al: Transforming growth factor-beta 1 induces intestinal myofibroblast differentiation and modulates their migration. World J Gastroenterol. 15:1431–1442. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Andersson-Sjöland A, Erjefält JS, Bjermer L, Eriksson L and Westergren-Thorsson G: Fibrocytes are associated with vascular and parenchymal remodelling in patients with obliterative bronchiolitis. Respir Res. 10:1032009.PubMed/NCBI

17 

Tahara E: Abnormal growth factor/cytokine network in gastric cancer. Cancer Microenviron. 1:85–91. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Yoshida K, Yokozaki H, Niimoto M, Ito H, Ito M and Tahara E: Expression of TGF-beta and procollagen type I and type III in human gastric carcinomas. Int J Cancer. 44:394–398. 1989. View Article : Google Scholar : PubMed/NCBI

19 

Wells RG: Fibrogenesis. V: TGF-beta signaling pathways. Am J Physiol Gastrointest Liver Physiol. 279:G845–850. 2000.PubMed/NCBI

20 

Blobe GC, Schiemann WP and Lodish HF: Role of transforming growth factor beta in human disease. N Engl J Med. 342:1350–1358. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Zavadil J and Böttinger EP: TGF-beta and epithelial-to-mesenchymal transitions. Oncogene. 24:5764–5774. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Zhang XY, Pettengell R, Nasiri N, Kalia V, Dalgleish AG and Barton DP: Characteristics and growth patterns of human peritoneal mesothelial cells: comparison between advanced epithelial ovarian cancer and non-ovarian cancer sources. J Soc Gynecol Investig. 6:333–340. 1999.

24 

Jayne DG, Perry SL, Morrison E, Farmery SM and Guillou PJ: Activated mesothelial cells produce heparin-binding growth factors: implications for tumour metastases. Br J Cancer. 82:1233–1238. 2000.PubMed/NCBI

25 

Kajiyama H, Shibata K, Ino K, Nawa A, Mizutani S and Kikkawa F: Possible involvement of SDF-1alpha/CXCR4-DPPIV axis in TGF-beta1-induced enhancement of migratory potential in human peritoneal mesothelial cells. Cell Tissue Res. 330:221–229. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Aroeira LS, Aguilera A, Sánchez-Tomero JA, et al: Epithelial to mesenchymal transition and peritoneal membrane failure in peritoneal dialysis patients: pathologic significance and potential therapeutic interventions. J Am Soc Nephrol. 18:2004–2013. 2007. View Article : Google Scholar

27 

Nishimura S, Chung YS, Yashiro M, Inoue T and Sowa M: Role of alpha 2 beta 1- and alpha 3 beta 1-integrin in the peritoneal implantation of scirrhous gastric carcinoma. Br J Cancer. 74:1406–1412. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Yung S, Li FK and Chan TM: Peritoneal mesothelial cell culture and biology. Perit Dial Int. 26:162–173. 2006.PubMed/NCBI

29 

Yashiro M, Chung YS, Nishimura S, Inoue T and Sowa M: Peritoneal metastatic model for human scirrhous gastric carcinoma in nude mice. Clin Exp Metastasis. 14:43–54. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Yashiro M, Chung YS, Inoue T, et al: Hepatocyte growth factor (HGF) produced by peritoneal fibroblasts may affect mesothelial cell morphology and promote peritoneal dissemination. Int J Cancer. 67:289–293. 1996. View Article : Google Scholar : PubMed/NCBI

31 

Yang S, Pham LK, Liao CP, Frenkel B, Reddi AH and Roy-Burman P: A novel bone morphogenetic protein signaling in heterotypic cell interactions in prostate cancer. Cancer Res. 68:198–205. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Yasumoto K, Koizumi K, Kawashima A, et al: Role of the CXCL12/CXCR4 axis in peritoneal carcinomatosis of gastric cancer. Cancer Res. 66:2181–2187. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2012
Volume 41 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tsukada T, Fushida S, Harada S, Yagi Y, Kinoshita J, Oyama K, Tajima H, Fujita H, Ninomiya I, Fujimura T, Fujimura T, et al: The role of human peritoneal mesothelial cells in the fibrosis and progression of gastric cancer . Int J Oncol 41: 476-482, 2012
APA
Tsukada, T., Fushida, S., Harada, S., Yagi, Y., Kinoshita, J., Oyama, K. ... Ohta, T. (2012). The role of human peritoneal mesothelial cells in the fibrosis and progression of gastric cancer . International Journal of Oncology, 41, 476-482. https://doi.org/10.3892/ijo.2012.1490
MLA
Tsukada, T., Fushida, S., Harada, S., Yagi, Y., Kinoshita, J., Oyama, K., Tajima, H., Fujita, H., Ninomiya, I., Fujimura, T., Ohta, T."The role of human peritoneal mesothelial cells in the fibrosis and progression of gastric cancer ". International Journal of Oncology 41.2 (2012): 476-482.
Chicago
Tsukada, T., Fushida, S., Harada, S., Yagi, Y., Kinoshita, J., Oyama, K., Tajima, H., Fujita, H., Ninomiya, I., Fujimura, T., Ohta, T."The role of human peritoneal mesothelial cells in the fibrosis and progression of gastric cancer ". International Journal of Oncology 41, no. 2 (2012): 476-482. https://doi.org/10.3892/ijo.2012.1490