Liver epithelial cells inhibit proliferation and invasiveness of hepatoma cells

  • Authors:
    • Kuo-Shyang Jeng
    • Chi-Juei Jeng
    • Wen-Juei Jeng
    • I-Shyan Sheen
    • Shih-Yun Li
    • Zih-Hang Hung
    • Hsin-I Hsiau
    • Ming-Che Yu
    • Chiung-Fang Chang
  • View Affiliations

  • Published online on: December 8, 2015     https://doi.org/10.3892/or.2015.4478
  • Pages: 1622-1628
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is a worldwide malignancy with poor prognosis. Liver progenitors or stem cells could be a potential therapy for HCC treatment since they migrate toward tumors. Rat liver epithelial (RLE) cells have both progenitor and stem cell-like properties. Therefore, our study elucidated the therapeutic effect of RLE cells in rat hepatoma cells. RLE cells were isolated from 10-day old rats and characterized for stem cell marker expression. RLE cells and rat hepatoma cells (H4-IIE-C3 cells) were co-cultured and divided into four groups with different ratios of RLE and hepatoma cells. Group A had only rat hepatoma cells as a control group. The ratios of rat hepatoma and RLE cells in group B, C and D were 5:1, 1:1 and 1:5, respectively. Effective inhibition of cell proliferation and migration was found in group D when compared to group A. There was a significant decrease in Bcl2 expression and increase in late apoptosis of rat hepatoma cells when adding more RLE cells. RLE cells reduced cell proliferation and migration of rat hepatoma cells. These results suggested that RLE cells could be used as a potential cell therapy.

Introduction

Hepatocellular carcinoma (HCC) is one of the leading malignancies worldwide, particularly in Asia (13). The treatment outcomes for those who are not candidates for resection or transplantation are usually miserable. Even after surgery, the long-term prognosis is generally unsatisfactory due to the high recurrence rate and the lack of effective systemic therapy (4,5). The benefit of the present target therapy remains limited (68). Therefore, it is urgent to develop a novel therapy for these patients.

Progenitor and/or stem cells could be potential therapeutic agents for HCC. Bone marrow-derived mesenchymal stem cells may inhibit hepatoma cell growth and their invasiveness (913). However, to obtain such cells from bone marrow is somewhat difficult in clinical practice. Therefore, it would be better to use liver progenitor cells to treat liver diseases.

Rat liver epithelial (RLE) cells from WB-F344 rats had the characteristics of progenitor or stem cells (14). They could reduce tumorigenity of hepatoma CBRH-7919 cells by TGFβ/Smad signaling. Peters et al found that co-culture of primary rat hepatocytes and RLE cells were able to enhance interleukin-6-induced acute phase protein responses (15). However, to use RLE cells as a weapon to treat HCC, two main points still need to be elucidated. One is whether these RLE cells inhibit tumor growth, and the other is whether they inhibit tumor invasiveness. Therefore, our study was focussed on the understanding of cell-to-cell interaction between RLE and HCC cells.

Materials and methods

Rat hepatoma cells

The rat hepatoma cell line H4-IIE-C3 was obtained from the Bioresource Collection and Research Center (Hsinchu, Taiwan). Cells were cultured in the Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% (v/v) fetal bovine serum (FBS), 1% penicillin/streptomycin (Gibco, Carlsbad, CA, USA) at 37°C in a humidified 5% CO2 incubator, and were routinely sub-cultured with 0.05% trypsin in phosphate-buffer at 80–90% confluence.

Isolation and culture of RLE cells

Pathogen-free Fisher (F344) rats were purchased from the National Laboratory Animal Center (Taipei, Taiwan). Rats were housed at Far Eastern Memorial Hospital. All animal study was performed in accordance within the guide for the care and use of laboratory animals and with the approval protocols of the Institutional Animal Care and Use Committee in the Far Eastern Memorial Hospital (FEMH; IACUC Approval No: 99-1-43-C1). Ten day old Fisher F344 rats were used to isolate RLE cells. Liver pieces were incubated in a DMEM/F12 containing 10 mM HEPES (both from Gibco), 1 mg/ml type IV collagenase (Sigma, St. Louis, MO, USA) and 1% penicillin/streptomycin at 37°C for 20 min. RLE cells were plated on collagen I-coated culture dishes incubated at 37°C in humidity incubator with 5% CO2. Cells were grown in a stem cell medium containing DMEM/F12, 2% FBS, 10 mM HEPES, 0.1% ITS Premix (Corning, Corning, NY, USA), 1×10−7 M dexamethasone (Sigma), 10 ng/ml human stem cell factor (SCF; eBioscience, San Diego, CA, USA), 20 ng/ml epidermal growth factor (EGF) (Sigma, St. Louis, MO, USA) and penicillin/streptomycin.

Western blotting

RLE cell lysates were collected by a lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% sodium deoxycholate, protease inhibitors, pH 7.5). Total protein (10 mg) was separated in SDS-PAGE and transferred onto a polyvinylidene fluoride (PVDF) membrane (Millipore, Billerica, MA, USA). Blots were blocked and incubated with primary antibodies against albumin, CK19 (10712-1-AP), EpCAM (21050-1-AP) (both from Proteintech, Chicago, IL, USA) and β-actin as a loading control (Sigma). The appropriate HRP-conjugated secondary antibodies were used and enhanced chemiluminescence (ECL) detection system (Millipore) was employed to visualize the proteins. Images were collected by ImageQuant™ LAS 4000 (GE Healthcare Life Sciences, UK).

Co-culture of hepatoma and RLE cells

Cell culture inserts (Millipore) were used to set-up a separated co-culture system. Rat hepatoma (5×104) (H4-IIE-C3) cells were placed in the 24-well plate at the bottom and RLE cells were in the culture inserts. There were 4 groups for the co-culture systems. The control group (group A) only had H4-IIE-C3 cells without any RLE cells. In group B-D, different numbers of RLE cells: 1×104, 5×104 and 25×104 were placed, respectively. The ratios of RLE to H4-IIE-C3 cells were 1:5; 1:1 and 5:1, respectively.

Cell proliferation assay

Rat hepatoma H4-IIE-C3 cells were separately co-cultured with different amount of RLE cells as above (group A-D). Rat hepatoma cells from each group were harvested after 24, 48, 72 and 96 h following the co-culture, respectively. The proliferation of rat hepatoma cells was measured by the WST-1 cell proliferation assay (Roche, Mannheim, Germany). The optical density (OD) values at 450/690 nm were measured by an ELISA reader (Bio-Rad, Hercules, CA, USA).

Flow cytometry

RLE cells were stained with stem cell marker Thy-1 PE (BD Pharmingen, San Jose, CA, USA). For apoptosis assay, rat hepatoma cells were stained with FITC/Annexin V apoptosis detection kit I (BD Pharmingen) after a 3-day co-culture. All procedures were followed by the manufacturer's instructions. The data were collected on a FACSCalibur (BD Biosciences) and analyzed by FlowJo software (Tree Star, Inc., Ashland, OR, USA).

Migration assay

Transwell inserts (8-µm-pore) were used for the cell migration assay (Corning Inc., Tewksbury, CA, USA). In all groups, H4-IIE-C3 cells (5×104) were placed on the cell inserts in a serum-free DMEM and RLE cells were cultured in stem cell medium in a 24-well plate. Group A had only H4-IIE-C3 cells without RLE cells. The cell number of RLE cells in group B-D was 1×104, 5×104 and 25×104, respectively. The migrated rat hepatoma cells were evaluated after 24-h post-incubation at 37°C. The migrated cells were fixed with 10% formaldehyde and washed with phosphate-buffered saline (PBS). The cells were then stained with 0.4% Giemsa (Sigma) for 2 h and washed with sterile ddH2O. Images (magnification, ×100) were collected under a Leica microscope (Leica Microsystems, Wetzlar, Germany).

RNA isolation and quantitative real-time PCR

Total RNA was isolated from rat hepatoma cells H4-IIE-C3 by the innuPREP RNA Mini kit (Analytik Jena, Jena, Germany) according to the manufacture's protocol. The total RNA was reversely transcripted to cDNA by a High-Capacity cDNA Reverse Transcription kit (Applied Biosystems, Foster City, CA, USA). The mRNA expression was analyzed by a real-time PCR Roche LightCycler 480 (Roche Applied Science, Mannheim, Germany). For real-time PCR, procedures were as follows: hot start at 95°C for 1 min, followed by 45 cycles of denaturing at 95°C for 10 sec, annealing at 58°C for 5 sec and extension at 72°C for 20 sec. PCR products were detected using 2% agarose gel to confirm the expected sizes. All primer sequences for quantitative real-time PCR analysis were: Bcl2 forward, 5′-CGA CTT TGC AGA TGT CCA-3′ and Bcl2 reverse, 5′-ATG CCG GTT CAG GTA CTC AG-3′; Bax forward, 5′-GAG AGG ATG GCT GGG GAG AC-3′ and Bax reverse, 5′-TGA GTG AGG CAG TGA GGA CT-3′; GAPDH forward, 5′-CAC CAC CAA CTG CTT AG-3′ and GAPDH reverse, 5′-CTT CAC CAC CTT CTT GAT G-3′. Gene expression was analyzed after normalization to control gene GAPDH.

Statistical analysis

Comparisons among groups were performed using SPSS (SPSS, Inc., Chicago, IL, USA). All the data are reported as mean ± SD. Comparisons between different groups for each point were performed using the one-way analysis of variance (ANOVA; and Kruskal-Wallis test), and multivariate analysis. All tests were two-tailed, and p<0.05 was considered to indicate a statistically significant result.

Results

Characterization of RLE cells

Isolated RLE cells started to form a colony within 2 days and the cells were confluent after a 5-day culture (Fig. 1A and B). The morphology of RLE cells changed after a few passages, notably when comparing the 1st and 4th passage (Fig. 1C and D). Cells from the 1st passage were round, whereas the 4th passage cells became fibroblast-like, which suggested that cells became unhealthy after the 4th generation passage. Therefore, RLE cells only from the 2nd and 3rd passages were used in the present study.

In addition, 75% of RLE cells expressed stem cell marker Thy-1 (Fig. 2A). Notably, stem cell markers CK19 and EpCAM as well as hepatocyte marker albumin were detected in RLE cells (Fig. 2B).

RLE cells reduce cell proliferation of rat hepatoma cells

There was a decreased tendency of cell proliferation in rat hepatoma cells when the RLE cells increased (Fig. 3). However, there was no difference between group A and D; B and C; or C and D. A significant difference between group B and D (p=0.049) at day 4 was found, which showed that an increased cell number of RLE cells reduced the cell proliferation in rat hepatoma cells.

RLE cells increased apoptosis of rat hepatoma cells

There were no differences in early apoptosis (Annexin V+/PI) among the groups. Group A (hepatoma cells only) had 3% early apoptosis cells and 16% late apoptotic cells. However, the percentages of late apoptotic cells (Annexin V+/PI+) increased among the other three groups, from 10% (group B), 14% (group C) to 16% (group D) (Fig. 4). This shows that the apoptotic rates of rat hepatoma cells increased by adding more RLE cells.

RLE cells reduced cell migration of rat hepatoma cells

The migration of rat hepatoma was examined at the early time point (day 2) since RLE cells could reduce cell proliferation at the late time point (day 4). There was a significant decrease of the number of the migrated rat hepatoma cells in comparison between group A and D (p=0.029) (Fig. 5). In addition, there was a decreased trend of migrated rat hepatoma cells when RLE cell number increased. Therefore, RLE cells inhibited the migration of rat hepatoma cells.

Decreased Bcl2 expression in rat hepatoma cells with increasing RLE cells

When comparing group A (rat hepatoma cells only) to group D (rat hepatoma cells: RLE cells=1:5), there was a significant decrease of the survival gene Bcl2 (p=0.005) (Fig. 6A). There seemed to be an increase of apoptotic gene Bax expression in group D. However, it was not statistically significant (Fig. 6B). It suggested that increasing RLE cells can reduce the survival of rat hepatoma cells.

Discussion

Rat liver epithelial (RLE) cells isolated from adult and newborn rats have similar oval cell-like properties (16). In our study, RLE cells were isolated from new-born rats and they could express progenitor cell or stem cell makers such as Thy-1, CK19, EpCAM and albumin. Thy-1 and CK19 are also expressed in human fetal livers (17). Hepatic progenitor cells express the markers EpCAM, CK7, CK19, α-fetoprotein (AFP) and Thy-1 (1820). It suggests that RLE cells have similar properties and characteristics as hepatocyte progenitor cells.

Two important points were found in the present study. First, RLE cells reduced the cell proliferation and survival of hepatoma cells. Second, RLE cells could also inhibit the migration of tumor cells. Both effects significantly enhanced when RLE cell number was increased. RLE cells induced apoptosis and reduced the survival of rat hepatoma cells due to downregulation in Bcl2. Bcl2 is a survival factor and highly expressed in hepatocellular carcinoma (HCC) cells. Therefore, targeting Bcl2 could induce cell death in tumors (21). It is also true that hepatocyte precursors increased apoptosis of rat hepatoma cells by decreasing Bcl2 and c-Myc (14).

In addition, RLE cells were able to reduce the migration of rat hepatoma cells. Tumor migration is one characteristic of cancer invasiveness (22,23). It could come from cytokines or chemokines made by RLE cells since the co-culture system was used without cell-to-cell contact. Previous study showed cytokine IL-6 was found to increase fibrinogen and decrease albumin secretion when co-culturing epithelial cells with hepatocytes (15). It is possible that IL-6 participated in the reduction of cell migration. TGFβ and its receptors (TBRI and TBRII) are involved in tumor cell invasion through epithelial-mesenchymal transition (EMT). Therefore, increased TGFβ receptor and Smad expression was found in rat hepatoma cells when increasing hepatocytic precursor cells (14).

In other studies, rat liver stem cells may selectively migrate to rat HCC due to chemoattractants or factors such as VEGF, PDGF, TGF-β, MCP-1, IL-8, TNF-α, IL-1β, IL-6, SDF-1 or HGF (2426). Therefore, hepatocyte progenitor cells could be used for cell therapy in HCC (27).

Stem cells for cancer therapy have been reported. Human-induced pluripotent stem cells (iPS) which can differentiate into neural stem cells (NSC) may be used as a cellular delivery vehicle for cancer gene therapy (28). It remains complex that mesenchymal stem cells (MSCs) either promote or inhibit different tumors (Table I). MSCs derived from bone marrow have characteristics in tumor suppression (1012,2936). On the contrary, some found bone marrow-derived MSCs promote tumor growth (3740). However, MSCs could act as a therapeutic tool and gene carrier for liver fibrosis and HCC (41,42). Whether stem cells or progenitor cells inhibit or promote tumor growth remains controversial, depending upon the stem cell origin and the difference among neoplasms.

Table I

Mesenchymal stem cells either suppress or promote tumor growth.

Table I

Mesenchymal stem cells either suppress or promote tumor growth.

AuthorNeoplasmEffectRefs.
Ho et alGliomaSuppression10
Lu et alMurine hepatoma H22
Lymphoma (YAC-1 and EL-4)
Rat insulinoma INS-1 cell line
Inhibition29
Ramasamy et alTumor cell line of hematopoietic (BV173)Inhibition30
Khakoo et alKaposi's sarcomaInhibition31
Elzaouk et alMouse melanomaAntitumor, IL-1232
Li et alHCCEnhanced tumor growth but inhibition invasiveness and metastasis33
Wang et alEsophageal cancerDeclining tumorigenicity34
Long et alHepG2 cell lineGrowth inhibitory11
Bruno et alHepG2 hepatoma Kaposi's sarcoma
Skov-3 ovarian tumor cell lines
Inhibit in vitro cell growth and survival of different tumor cell lines and in vivo tumor growth12
Qiao et alH7402 and HepG2 human liver cell lineInhibit proliferation, colony-forming ability and oncogene expression both in vitro and in vivo35
Ganta et alBreast cancer Antiproliferative36
Zhu et alF6 tumor cell line
SW480 tumor cell line
Favor growth37
Djouad et alMelanoma tumor cellsFavor growth38
Jung et alProstate cancerPromote metastasis39
Karnoub et alBreast cancerPromote metastasis40

In conclusion, RLE cells were able to inhibit proliferation and invasiveness of rat hepatoma cells. Therefore, RLE cells have a high potential for cell therapy in hepatoma cells.

Abbreviations:

HCC

hepatocellular carcinoma

RLE

rat liver epithelial

AFP

α-fetoprotein

EMT

epithelial-mesenchymal transition

iPS

induced pluripotent stem

NSC

neural stem cells

MSC

mesenchymal stem cells

PI

propidium iodide

Acknowledgments

We greatly appreciate the support and equipment received from Core Laboratory of Far Eastern Memorial Hospital. We also thank Miss Szu-Hua Wu and Fang-Yu Chang for their assistance. The present study was supported by grants from the Far Eastern Memorial Hospital (FEMH-2012-SCRM-A-001), the Taiwan National Science Council (NSC 100-2314-B-418-004-MY3) and the Ministry of Science and Technology, R.O.C (MOST 104-2314-B-418-018).

References

1 

Venook AP, Papandreou C, Furuse J and de Guevara LL: The incidence and Epidemiology of hepatocellular carcinoma: A global and regional perspective. Oncologist. 15(Suppl 4): S5–S13. 2010. View Article : Google Scholar

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Arzumanyan A, Reis HM and Feitelson MA: Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat Rev Cancer. 13:123–135. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Fonseca AL and Cha CH: Hepatocellular carcinoma: A comprehensive overview of surgical therapy. J Surg Oncol. 110:712–719. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Raza A and Sood GK: Hepatocellular carcinoma review: Current treatment, and evidence-based medicine. World J Gastroenterol. 20:4115–4127. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Frenette C and Gish R: Targeted systemic therapies for hepatocellular carcinoma: Clinical perspectives, challenges and implications. World J Gastroenterol. 18:498–506. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Wei Z, Doria C and Liu Y, Wei Y, Doria C and Liu Y: Targeted therapies in the treatment of advanced hepatocellular carcinoma. Clin Med Insights Oncol. 7:87–102. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Jeng KS, Chang CF, Jeng WJ, Sheen IS and Jeng CJ: Heterogeneity of hepatocellular carcinoma contributes to cancer progression. Crit Rev Oncol Hematol. 94:337–347. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Bayo J, Marrodán M, Aquino JB, Silva M, Garcia MG and Mazzolini G: The therapeutic potential of bone marrow-derived mesenchymal stromal cells on hepatocellular carcinoma. Liver Int. 34:330–342. 2014. View Article : Google Scholar

10 

Ho IA, Toh HC, Ng WH, Teo YL, Guo CM, Hui KM and Lam PY: Human bone marrow-derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis. Stem Cells. 31:146–155. 2013. View Article : Google Scholar

11 

Long X, Matsumoto R, Yang P and Uemura T: Effect of human mesenchymal stem cells on the growth of HepG2 and Hela cells. Cell Struct Funct. 38:109–121. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Bruno S, Collino F, Deregibus MC, Grange C, Tetta C and Camussi G: Microvesicles derived from human bone marrow mesenchymal stem cells inhibit tumor growth. Stem Cells Dev. 22:758–771. 2013. View Article : Google Scholar

13 

Zhang L, Su XS, Ye JS, Wang YY, Guan Z and Yin YF: Bone marrow mesenchymal stem cells suppress metastatic tumor development in mouse by modulating immune system. Stem Cell Res Ther. 6:452015. View Article : Google Scholar : PubMed/NCBI

14 

Li WQ, Li YM, Guo J, Liu YM, Yang XQ, Ge HJ, Xu Y, Liu HM, He J and Yu HY: Hepatocytic precursor (stem-like) WB-F344 cells reduce tumorigenicity of hepatoma CBRH-7919 cells via TGF-β/Smad pathway. Oncol Rep. 23:1601–1607. 2010.PubMed/NCBI

15 

Peters SJ, Vanhaecke T, Papeleu P, Rogiers V, Haagsman HP and van Norren K: Co-culture of primary rat hepatocytes with rat liver epithelial cells enhances interleukin-6-induced acute-phase protein response. Cell Tissue Res. 340:451–457. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Tsao MS, Smith JD, Nelson KG and Grisham JW: A diploid epithelial cell line from normal adult rat liver with phenotypic properties of ʻovalʼ cells. Exp Cell Res. 154:38–52. 1984. View Article : Google Scholar : PubMed/NCBI

17 

Masson NM, Currie IS, Terrace JD, Garden OJ, Parks RW and Ross JA: Hepatic progenitor cells in human fetal liver express the oval cell marker Thy-1. Am J Physiol Gastrointest Liver Physiol. 291:G45–G54. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Petersen BE, Goff JP, Greenberger JS and Michalopoulos GK: Hepatic oval cells express the hematopoietic stem cell marker Thy-1 in the rat. Hepatology. 27:433–445. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Yovchev MI, Grozdanov PN, Joseph B, Gupta S and Dabeva MD: Novel hepatic progenitor cell surface markers in the adult rat liver. Hepatology. 45:139–149. 2007. View Article : Google Scholar

20 

Hao PP, Lee MJ, Yu GR, Kim IH, Cho YG and Kim DG: Isolation of EpCAM+/CD133 hepatic progenitor cells. Mol Cells. 36:424–431. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Ni Z, Wang B, Dai X, Ding W, Yang T, Li X, Lewin S, Xu L, Lian J and He F: HCC cells with high levels of Bcl2 are resistant to ABT-737 via activation of the ROS-JNK-autophagy pathway. Free Radic Biol Med. 70:194–203. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Friedl P and Wolf K: Tumour-cell invasion and migration: Diversity and escape mechanisms. Nat Rev Cancer. 3:362–374. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Stivarou T and Patsavoudi E: Extracellular molecules involved in cancer cell invasion. Cancers. 7:238–265. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, Charbord P and Domenech J: The in vitro migration capacity of human bone marrow mesenchymal stem cells: Comparison of chemokine and growth factor chemotactic activities. Stem Cells. 25:1737–1745. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Pietras K, Pahler J, Bergers G and Hanahan D: Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med. 5:e192008. View Article : Google Scholar : PubMed/NCBI

26 

Watnick RS: The role of the tumor microenvironment in regulating angiogenesis. Cold Spring Harb Perspect Med. 2:a0066762012. View Article : Google Scholar : PubMed/NCBI

27 

Zhong XG, He S, Yin W, Deng JY and Cheng B: Selective tropism of liver stem cells to hepatocellular carcinoma in vivo. World J Gastroenterol. 13:3886–3891. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Yang J, Lam DH, Goh SS, Lee EX, Zhao Y, Tay FC, Chen C, Du S, Balasundaram G, Shahbazi M, et al: Tumor tropism of intravenously injected human-induced pluripotent stem cell-derived neural stem cells and their gene therapy application in a metastatic breast cancer model. Stem Cells. 30:1021–1029. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Lu YR, Yuan Y, Wang XJ, Wei LL, Chen YN, Cong C, Li SF, Long D, Tan WD, Mao YQ, et al: The growth inhibitory effect of mesenchymal stem cells on tumor cells in vitro and in vivo. Cancer Biol Ther. 7:245–251. 2008. View Article : Google Scholar

30 

Ramasamy R, Lam EW, Soeiro I, Tisato V, Bonnet D and Dazzi F: Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth. Leukemia. 21:304–310. 2007. View Article : Google Scholar

31 

Khakoo AY, Pati S, Anderson SA, Reid W, Elshal MF, Rovira II, Nguyen AT, Malide D, Combs CA, Hall G, et al: Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med. 203:1235–1247. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Elzaouk L, Moelling K and Pavlovic J: Anti-tumor activity of mesenchymal stem cells producing IL-12 in a mouse melanoma model. Exp Dermatol. 15:865–874. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Li GC, Ye QH, Xue YH, Sun HJ, Zhou HJ, Ren N, Jia HL, Shi J, Wu JC, Dai C, et al: Human mesenchymal stem cells inhibit metastasis of a hepatocellular carcinoma model using the MHCC97-H cell line. Cancer Sci. 101:2546–2553. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Wang Y, Fan H, Zhou B, Ju Z, Yu L, Guo L, Han J and Lu S: Fusion of human umbilical cord mesenchymal stem cells with esophageal carcinoma cells inhibits the tumorigenicity of esophageal carcinoma cells. Int J Oncol. 40:370–377. 2012.

35 

Qiao L, Xu Z, Zhao T, Zhao Z, Shi M, Zhao RC, Ye L and Zhang X: Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res. 18:500–507. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Ganta C, Chiyo D, Ayuzawa R, Rachakatla R, Pyle M, Andrews G, Weiss M, Tamura M and Troyer D: Rat umbilical cord stem cells completely abolish rat mammary carcinomas with no evidence of metastasis or recurrence 100 days post-tumor cell inoculation. Cancer Res. 69:1815–1820. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Zhu W, Xu W, Jiang R, Qian H, Chen M, Hu J, Cao W, Han C and Chen Y: Mesenchymal stem cells derived from bone marrow favor tumor cell growth in vivo. Exp Mol Pathol. 80:267–274. 2006. View Article : Google Scholar

38 

Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, Noël D and Jorgensen C: Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood. 102:3837–3844. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Jung Y, Kim JK, Shiozawa Y, Wang J, Mishra A, Joseph J, Berry JE, McGee S, Lee E, Sun H, et al: Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis. Nat Commun. 4:17952013. View Article : Google Scholar : PubMed/NCBI

40 

Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R and Weinberg RA: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Aquino JB, Bolontrade MF, García MG, Podhajcer OL and Mazzolini G: Mesenchymal stem cells as therapeutic tools and gene carriers in liver fibrosis and hepatocellular carcinoma. Gene Ther. 17:692–708. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Li GC, Ye QH, Dong QZ, Ren N, Jia HL and Qin LX: Mesenchymal stem cells seldomly fuse with hepatocellular carcinoma cells and are mainly distributed in the tumor stroma in mouse models. Oncol Rep. 29:713–719. 2013.

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jeng K, Jeng C, Jeng W, Sheen I, Li S, Hung Z, Hsiau H, Yu M and Chang C: Liver epithelial cells inhibit proliferation and invasiveness of hepatoma cells. Oncol Rep 35: 1622-1628, 2016
APA
Jeng, K., Jeng, C., Jeng, W., Sheen, I., Li, S., Hung, Z. ... Chang, C. (2016). Liver epithelial cells inhibit proliferation and invasiveness of hepatoma cells. Oncology Reports, 35, 1622-1628. https://doi.org/10.3892/or.2015.4478
MLA
Jeng, K., Jeng, C., Jeng, W., Sheen, I., Li, S., Hung, Z., Hsiau, H., Yu, M., Chang, C."Liver epithelial cells inhibit proliferation and invasiveness of hepatoma cells". Oncology Reports 35.3 (2016): 1622-1628.
Chicago
Jeng, K., Jeng, C., Jeng, W., Sheen, I., Li, S., Hung, Z., Hsiau, H., Yu, M., Chang, C."Liver epithelial cells inhibit proliferation and invasiveness of hepatoma cells". Oncology Reports 35, no. 3 (2016): 1622-1628. https://doi.org/10.3892/or.2015.4478