Human mesenchymal stem cells enhance cancer cell proliferation via IL-6 secretion and activation of ERK1/2

  • Authors:
    • Agmal Scherzad
    • Magdalena Steber
    • Thomas Gehrke
    • Kristen Rak
    • Katrin Froelich
    • Philipp Schendzielorz
    • Rudolf Hagen
    • Norbert Kleinsasser
    • Stephan Hackenberg
  • View Affiliations

  • Published online on: May 18, 2015     https://doi.org/10.3892/ijo.2015.3009
  • Pages: 391-397
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human mesenchymal stem cells (hMSC) are frequently used in tissue engineering. Due to their strong tumor tropism, hMSC seem to be a promising vehicle for anticancer drugs. However, interactions between hMSC and cancer are ambiguous. Particularly the cytokines and growth factors seem to play an important role in cancer progression and metastasis. The present study evaluated the effects of hMSC on head and neck squamous cell carcinoma (HNSCC) cell lines (FaDu and HLaC78) in vitro. hMSC released several cytokines and growth factors. FaDu and HLaC78 showed a significant enhancement of cell proliferation after cultivation with hMSC-conditioned medium as compared to control. This proliferation improvement was inhibited by the addition of anti-IL-6. The western blot showed an activation of Erk1/2 in FaDu and HLaC78 by hMSC-conditioned medium. HNSCC cell lines expressed EGFR. The current study confirms the importance of cytokines secreted by hMSC in cancer biology. Especially IL-6 seems to play a key role in cancer progression. Thus, the use of hMSC as a carrier for cancer therapy must be discussed critically. Future studies should evaluate the possibility of generating genetically engineered hMSC with, for example, the absence of IL-6 secretion.

Introduction

Human mesenchymal stem cells (hMSC) are undifferentiated cells capable of self-renewal and proliferation (1). Under certain conditions hMSC are able to differentiate into diverse mesenchymal cell types, e.g., osteocytes, adipocytes and chondrocytes (2). Due to their ability to migrate as well as the capability to differentiate, hMSC play an important role in the preservation and re-creation of tissue integrity. Inflammation and tissue damage are strong chemoattractants for hMSC, similar to the recruitment of leucocytes into inflamed tissue sites (35). Tissue-specific homing of leukocytes depends on cytokines, tissue-specific adhesion molecules, and homing receptors (6). Von Lüttichau and colleagues demonstrated functional expression of several chemokine receptors on primary isolated hMSC (7).

Cancer tissue exhibits similar cytokine patterns as inflamed tissue (8). Hence, there is a strong tropism of hMSC towards tumors (9). Furthermore, hMSC integrate into the tumor directly and build parts of the cancer microenvironment (9,10). Thus, several studies have suggested that hMSC would be an appropriate biological carrier, for example, for drug delivery therapy (11,12).

However, interactions between hMSC and cancer are ambiguous. Some studies show tumor progression and enhancement of the tumor metastatic potential by hMSC. This progression is induced by cell-cell contact as well as by the secretion of several cytokines and growth factors by hMSC such as interleukin-6 (IL-6), epidermal growth factor (EGF), vascular epidermal growth factor (VEGF), insulin-like growth factor (IGF)-1 or transforming growth factor (TGF)-β in a paracrine manner (1318). Some of these, especially EGF and IL-6, may play an important role in tumor progression and metastasis. The activation of mitogen-activated protein kinase (MAPK) pathways was shown to play an important role in cytokine-induced tumor progression (19,20). MAPK pathways link extracellular signals to the machinery that controls fundamental cellular processes (21). Especially the extracellular signal-regulated kinase (ERK)1/2 is activated by growth factors, which in turn is responsible for cell proliferation, differentiation, apoptosis and migration (21).

In a previous study, we were able to show an increase in tumor proliferation as well as motility in the presence of hMSC. Cancer cell lines showed resistance towards paclitaxel after co-cultivation with hMSC (18,22). The aim of the present study was to evaluate whether hMSC induce an activation of the MAPK pathway in vitro. Furthermore, the effect of IL-6 on cell proliferation was also investigated.

Materials and methods

hMSC isolation and culture

The cells were harvested and prepared from 5 voluntary patients with written informed consent from the Department of Orthopedics, Koenig-Ludwig-Haus. The study was approved by the Ethics Committee of the Medical Faculty of the University of Wuerzburg (12/06). Bone marrow was collected under aseptic conditions and cells were isolated according to the protocol described by Lee et al (23). Ficoll density-gradient centrifugation was used in order to isolate hMSC (30 min, 1,300 rpm, density=1,077 g/ml, Biochrom AG, Berlin, Germany). After collection of the cells from the interphase, a washing step with phosphate buffered saline (PBS) (Roche Diagnostics GmbH, Mannheim, Germany) containing 2% FCS followed. After centrifugation the cell pellet was resuspended in expansion medium (DMEM-EM), which was made of Dulbecco’s modified Eagle’s medium (DMEM) (Gibco Invitrogen, Karlsruhe, Germany) containing 10% FCS (Linaris, Wertheim-Bettingen, Germany), 1% penicillin and streptomycin (Sigma-Aldrich, Schnelldorf, Germany). Cells were incubated for 24 h at 37°C and 5% CO2. The medium was changed every other day. The morphology of hMSC was analyzed by inverted microscopy (Leica DMI 4000B Inverted Microscope, Leica Microsystems, Wetzlar, Germany).

Multidifferentiation capacity

Osteogenic differentiation was carried out in a 24-well plate (BD Falcon, Heidelberg, Germany) with 1×104 cells/well until 70% confluence was reached. The expansion medium DMEM-EM additionally contained 10−7 M dexamethasone, 10−3 M β-glycerophosphate and 2−4 M ascorbate-2-phosphate (all Sigma-Aldrich). The von Kossa method was used to show the presence of calcium mineral components. Adipogenic differentiation medium was made of DMEM-EM containing 10−7 M dexamethasone and 10−9 g/ml recombinant human insulin (both Sigma-Aldrich). Staining with Oil Red O confirmed the presence of intra-cellular lipid droplets.

For chondrogenic differentiation the pellet culture system was used. The cell pellets were cultured in a defined chondrogenic differentiation medium (Lonza, Basel, Switzerland) supplemented with 10−9 g/ml transforming growth factor-β3 (Sigma-Aldrich). Cells were cultivated for 3 weeks. Thereafter, the pellets were embedded in Optimal Cutting Temperature Paraffin (Tissue-Tek® O.C.TTM; Sakura Finetek, Zoeterwoude, The Netherlands). Cryosections were stained with Alcian blue to show the presence of glycosamineglycane.

Expression of cell surface markers

Flow cytometry was used in order to confirm surface antigen markers. hMSC (1×106) were incubated with anti-CD105, anti-CD90, anti-CD44 and anti-CD34 (all antibodies were purchased from BD Bioscience, Heidelberg, Germany). Cell surface analyses were performed by flow cytometry (FACSCanto™; BD Bioscience).

HNSCC cell line FaDu and HLaC78

The head and neck squamous carcinoma cell lines FaDu and HLaC78 were used (24,25). Cells were grown in DMEM-EM and cultured at 37°C with 5% CO2 in culture flasks. Every other day the medium was replaced. After reaching 70–80% confluence cells were trypsinized with 0.25% trypsin (Gibco Invitrogen), washed with PBS and seeded in new flasks or treatment wells. Experiments were performed using cells in the exponential growth phase. FaDu and HLaC78 were incubated with the supernatants of hMSC (hMSC-sup). The following experiments were carried out in order to evaluate the effects of cytokines released by hMSC on FaDu and HLaC78.

Cytokine analysis of hMSC-sup with the dot blot assay

The dot blot assay (RayBiotech Inc., Norcross, GA, USA) was used as a semi-quantitative method for determining hMSC cytokine secretion. After an incubation period of 48 h in DMEM without supplements, the supernatants were collected and investigated for the presence of cytokines. The assay was performed according to the manufacturer’s protocol. The labeled proteins were observed by enhanced chemiluminescence using detection buffer and exposure to X-ray film. The cytokines were represented as dots with different intensity and growth size.

Quantitative analysis of IL-6 by ELISA

Supernatants of hMSC were collected. IL-6 concentration was measured using the ELISA kit human IL-6 (Disclose SAS, Besancon Codex, France). All experiments were investigated in duplicate. The plate was read at 450 nm (Titertek Multiskan PLUS; Labsystems, Helsinki, Finland). IL-6 concentration (pg/ml) was ascertained by creating a standard curve using recombinant IL-6. DMEM without supplements served as the control.

Cell proliferation analysis

Cells (2×104) (FaDu and HLaC78) were incubated with hMSC-sup at 37°C with 5% CO2 for 4 days, while electronically counting the cell number each day (Casy® Technologies, Innovatis AG, Reutlingen, Germany). DMEM-EM served as the control (n=5).

Simultaneously, the effect of IL-6 on cell proliferation was evaluated by using the MTT assay. Cells were seeded at a density of 1×104 in a 96-well rounded bottom plate. After an incubation period of 24 h in DMEM-EM, hMSC-sup +/− anti-IL-6 500 ng/ml (R&D Systems, Wiesbaden-Nordenstadt, Germany) cells were washed with PBS. Then all plates were incubated with 100 μl of MTT solution (1 mg/ml) followed by 5 h of incubation at 37°C with 5% CO2. After removal of MTT, 100 μl of isopropanol was added for 1 h at 37°C with 5% CO2. The plate was read at a wavelength of 570 nm (Titertek Multiskan) (n=5).

Total RNA extraction, cDNA synthesis and real-time PCR

The expression of EGFR in FaDu and HLaC78 were investigated as follows: According to the manufacturer’s protocol, the TRIzol method was used in order to extract total RNA from cells. Next, the extracted total RNA was reverse transcribed to cDNA. For the quantification of gene expression, the SYBR Green PCR master mix kit (Applied Biosystems, Foster City, CA, USA) was used. The EGFR primer was purchased from Applied Biosystems (forward primer 5′-GCGTTCGGCACGGTGTATAA-3′ and reverse primer 5′-GGCTTTCGGAGATGTTGCTTC-3′). The conditions for gene amplification were as follows: 50°C for 2 min; 95°C for 10 min, and 40 cycles at 95°C for 15 sec and 60°C for 1 min. As an endogenous control, the GAPDH gene was used.

Western blotting

Cells (FaDu and HLaC78) were harvested by trypsinization and dissolved in RIPA buffer (PBS, containing 1% NP40, 0.5% sodium deoxycholate and 0.1% SDS), supplemented with 10 μg/ml phenylmethanesulfonyl fluoride (PMSF). Protein concentration was then determined. Equal amounts of total protein lysates were loaded on a 10% SDS-polyacrylamide gel and transferred by electroblotting to a polyvinylidene difluoride membrane. Blots were blocked for 1 h at room temperature with TBST (10 mM Tris, 150 mM NaCl, 0.05% Tween-20, pH 8.0), containing 5% nonfat dry milk. Afterwards, the membrane was incubated with primary antibody ERK1/2 (Cell Signaling Technology, Beverly, MA, USA) overnight at 4°C. Subsequently, the membrane was washed and incubated with a species-specific IgG secondary antibody for 1 h to visualize the specific bindings. The protein expression was detected with a chemiluminescence system (ECL, Amersham Biosciences, Freiburg, Germany), according to the manufacturer’s protocol.

Statistical analysis

All data were transferred to standard spreadsheets. Differences between groups were examined for significance by the Kruskal-Wallis test using GraphPad Prism 6.0 statistics software (GraphPad Software, Inc., San Diego, CA, USA). Differences were considered statistically significant when the P-value was <0.05.

Results

hMSC morphology and differentiation capability

hMSC exhibited a fibroblast-shaped morphology. They were able to differentiate into chondrocytes, adipocytes and osteocytes, which were confirmed by Alcian blue, Oil Red O and van Kossa staining. The flow cytometric analysis revealed the presence of typical surface markers. hMSC were positive for CD105, CD90 and CD44, and negative for CD34 (Figs. 1 and 2).

HNSCC cell line morphology before and after cultivation with hMSC-sup

Morphology of FaDu and HLaC78 was evaluated before and after cultivation with hMSC-sup for 48 h. Microscopy revealed no differences in cell morphology between each group for both cell lines.

Cytokine secretion by hMSC

The dot blot assay was used to evaluate different cytokines in the hMSC supernatants. hMSC supernatants contained a variety of cytokines responsible for pro- and anti-inflammation, chemotaxis, angiogenesis and growth factors. The dots of the following cytokines had a strong intensity: interleukin (IL)-3; IL-6, IL-8, IL-10, Growth regulated oncogene (GRO), GRO-α, Monocyte chemotactic protein (MCP)-1, Macrophage colony-stimulating factor, Stem cell factor, Tumor necrosis factor (TNF)-α and OncostatinM (Fig. 3 and Table I).

Table I

Cytokines map.

Table I

Cytokines map.

++ENA-78GCSFGM-CSFGROGRO-αI-309IL-1αIL-1β
++ENA-78GCSFGM-CSFGROGRO-αI-309IL-1αIL-1β
IL-2IL-3IL-4IL-5IL-6IL-7IL-8IL-10IL-12IL-13IL-15IFN-γ
IL-2IL-3IL-4IL-5IL-6IL-7IL-8IL-10IL-12IL-13IL-15IFN-γ
MCP-1MCP-2MCP-3MCSFMCDMIGMIP-1dRANTESSCFSDF-1TARCTGF-β1
MCP-1MCP-2MCP-3MCSFMCDMIGMIP-1dRANTESSCFSDF-1TARCTGF-β1
TNF-αTNF-βEGFIGF-1AngiogeninOncostatinMThrombopoietinVEGFPDGF-BBLeptin+
TNF-αTNF-βEGFIGF-1AngiogeninOncostatinMThrombopoietinVEGFPDGF-BBLeptin+
Quantitative analysis of IL-6

The secretion of IL-6 was quantified with the ELISA method. hMSC in monolayer culture released 260 pg/ml IL-6. DMEM without supplements as control did not contain IL-6 (Fig. 4).

Effects of hMSC on HNSCC proliferation +/− anti-IL-6

The cultivation of FaDu and HLaC78 with hMSC-sup induced a significant enhancement of cell proliferation compared to cultivation in DMEM-EM. The MTT assay confirmed the results of the cell counting. FaDu and HLaC78 showed a significant enhancement of cell proliferation. The impact of IL-6 on cell proliferation was investigated by the addition of anti-IL-6 into hMSC-sup. Anti-IL-6 induced the inhibition of cell proliferation. FaDu and HLaC78 showed a reduction in cell proliferation compared to cultivation with hMSC-sup without anti-IL-6 (Figs. 5 and 6).

Expression of EGFR in HNSCC

fGFR gene expression was assessed by real-time PCR in FaDu and HLaC78. Values were expressed using the ΔΔCt method to derive relative fold change. Both cell lines were positive for EGFR expression. The EGFR expression was almost 2-fold more in HLaC78 compared to FaDu (Fig. 7).

Activation of ERK1/2

The expression of ERK1/2 and the corresponding phosphorylated protein in FaDu and HLaC78 was investigated with the western blotting. Western blot analysis revealed expression of ERK1/2 as well as its phosphorylated protein after cultivation in hMSC-sup. The cultivation in DMEM-EM revealed total absence of the phosphorylated protein (Fig. 8).

Discussion

Solid tumors are embedded into a complex microenvironment, which includes several types of non-malignant cells, malignant cells as well as extracellular matrix. On one hand, cytokines produced by tumor cells alter the gene expression of stromal cells and their phenotypical properties. On the other hand, the cancer cells release growth factors, cytokines and proteases which enhance tumor growth (26,27). Cancer and stroma interact in an autocrine as well as paracrine manner. This implies the symbiotic relationship between cancer and cancer surrounding stroma. Several cytokines responsible for inflammation and angiogenesis are elevated in the serum of patients with HNSCC compared to the control group (28). IL-6, a multi-functional regulator of immune responses and hematopoiesis, influences the proliferation and invasion potential of head and neck cancer directly (29). IL-6 regulates a complex network of cytokines responsible for inflammation, growth factors as well as angiogenic proteins which lead to malignant and invasive tumor growth (30). In the current study, the cultivation of the HNSCC cell lines FaDu and HLaC78 with hMSC-sup resulted in proliferation enhancement of tumor cells. The dot blot of hMSC-sup revealed the secretion of various cytokines and growth factors by hMSC. The dots of several cytokines, especially IL-6, had a strong intensity.

The addition of anti-IL-6 counteracted these pro-mitotic effects of hMSC-sup. In the current study, a significant attenuation of HNSCC cell proliferation in the culture system treated with anti-IL-6 compared to cells treated with hMSC-sup was detected. This is an indirect marker for the impact of IL-6 on cancer progression. Furthermore, IL-6 can be responsible for the development of resistance towards anti-cancer drugs such as cisplatin (31). This was achieved by an increased expression of the cellular inhibitor of apoptosis 2 (cIAP-2) via IL-6. The effect was reversible by the addition of anti-IL-6 and anti-cIAP-2.

In the current study, the proliferation enhancement of FaDu and HLaC78 was induced via activation of ERK1/2. The Erk1/2 pathway is a critical signal transduction pathway in the mitogen-activated protein kinase family, and is closely associated with tumorigenesis and tumor progression (32). Erk1/2 targets different molecules that are responsible for cell growth, survival, adhesion, motility and differentiation (32,33). The activation of ERK1/2 via hMSC seems to be crucial for cell proliferation. IL-6 was shown by Shi et al to stimulate tumor growth by activation of Ras, Raf, MEK, and Erk1 and 2 (34). IL-6 induces different intracellular signaling cascades, e.g. JAK-STAT, MAPK and PI3K (35,36).

In malignant mammary carcinoma the activation of Erk1/2 has been associated with a poor prognosis (20). Due to their ability to be recruited by damaged tissues, inflammation and cancer, hMSC have been discussed as being a very promising vehicle for cancer treatment. However, many studies in this area do not consider the interactions between stem cells and cancer. There are studies showing a tumorigenic effect of hMSC (13,15,18), as well as reports on cancer inhibition by hMSC (37,38).

Karnoub et al were able to show that hMSC enhanced the metastatic potential of breast carcinoma cells by de novo secretion of RANTES, which represents a potent cytokine responsible for cancer cell motility, invasion and migration (13). The dot blot assay revealed the secretion of RANTES by hMSC. In a previous study by our group, the enhancement of cancer cell motility was demonstrated (22). Co-injection of hMSC and breast cancer cells resulted in cancer progression (15).

One important issue in cancer progression is the ability of hMSC to integrate into tumor stroma and differentiate into cancer-associated fibroblasts (CAF) (10,39,40). Long-term co-culture of hMSC with cancer supernatants resulted in a differentiation of hMSC into myofibroblasts (41). These CAF (differentiated hMSC) are able to release cytokines responsible for cancer growth and matrix remodeling (42). Furthermore, CAF are able to express proteins involved in lactate absorption, lactate oxidation and reduced glucose absorption (43). CAF sustain cancer cell survival by buffering and recycling products of anaerobic metabolism (43). Cancer-associated fibroblasts are involved in the initiation of cancer invasion (44).

In contrast to studies showing the tumor-supporting potential of hMSC, there are also studies indicating anti-cancer effects: As shown by Khakoo, the inhibition of Kaposi sarcoma was induced via suppression of Akt protein kinase by hMSC.

Qiao et al demonstrated inhibition of human hepatoma cell lines via the Wnt signaling pathway (37). Zhu et al presented the DKK-1 (dickkopf-1) protein as a negative regulator of the Wnt signaling pathway (38). In a study conducted by Qiao et al hMSC inhibited cancer cell growth by secretion of DKK-1 (45). The reason for these contradictory findings may be explained by the use of different stem cell sources as well as different donor ages. Thus, the use of stem cells as a carrier for cancer therapy must be critically discussed. A possible solution may be the use of genetically engineered stem cells with, for example, the absence of IL-6 secretion. Therefore, a challenging future mission will be to identify an optimal stem cell with enhanced tumor tropism and highly anti-tumorigenic potential.

References

1 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Bianco P, Riminucci M, Gronthos S and Robey PG: Bone marrow stromal stem cells: Nature, biology, and potential applications. Stem Cells. 19:180–192. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Coffelt SB, Marini FC, Watson K, Zwezdaryk KJ, Dembinski JL, LaMarca HL, Tomchuck SL, Honer zu Bentrup K, Danka ES, Henkle SL, et al: The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proc Natl Acad Sci USA. 106:3806–3811. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, Kearne M, Magner M and Isner JM: Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 85:221–228. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Malek S, Kaplan E, Wang JF, Ke Q, Rana JS, Chen Y, Rahim BG, Li M, Huang Q, Xiao YF, et al: Successful implantation of intravenously administered stem cells correlates with severity of inflammation in murine myocarditis. Pflugers Arch. 452:268–275. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Campbell JJ and Butcher EC: Chemokines in tissue-specific and microenvironment-specific lymphocyte homing. Curr Opin Immunol. 12:336–341. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Von Lüttichau I, Notohamiprodjo M, Wechselberger A, Peters C, Henger A, Seliger C, Djafarzadeh R, Huss R and Nelson PJ: Human adult CD34 progenitor cells functionally express the chemokine receptors CCR1, CCR4, CCR7, CXCR5, and CCR10 but not CXCR4. Stem Cells Dev. 14:329–336. 2005. View Article : Google Scholar

8 

Dvorak HF: Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 315:1650–1659. 1986. View Article : Google Scholar : PubMed/NCBI

9 

Kidd S, Caldwell L, Dietrich M, Samudio I, Spaeth EL, Watson K, Shi Y, Abbruzzese J, Konopleva M, Andreeff M, et al: Mesenchymal stromal cells alone or expressing interferon-beta suppress pancreatic tumors in vivo, an effect countered by anti-inflammatory treatment. Cytotherapy. 12:615–625. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, Andreeff M and Marini F: Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One. 4:e49922009. View Article : Google Scholar : PubMed/NCBI

11 

Huang Q, Liu XZ, Kang CS, Wang GX, Zhong Y and Pu PY: The anti-glioma effect of suicide gene therapy using BMSC expressing HSV/TK combined with overexpression of Cx43 in glioma cells. Cancer Gene Ther. 17:192–202. 2010. View Article : Google Scholar

12 

Cavarretta IT, Altanerova V, Matuskova M, Kucerova L, Culig Z and Altaner C: Adipose tissue-derived mesenchymal stem cells expressing prodrug-converting enzyme inhibit human prostate tumor growth. Mol Ther. 18:223–231. 2010. View Article : Google Scholar :

13 

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

14 

Lin WR, Brittan M and Alison MR: The role of bone marrow-derived cells in fibrosis. Cells Tissues Organs. 188:178–188. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Rhodes LV, Muir SE, Elliott S, Guillot LM, Antoon JW, Penfornis P, Tilghman SL, Salvo VA, Fonseca JP, Lacey MR, et al: Adult human mesenchymal stem cells enhance breast tumorigenesis and promote hormone independence. Breast Cancer Res Treat. 121:293–300. 2010. View Article : Google Scholar

16 

Kemp KC, Hows J and Donaldson C: Bone marrow-derived mesenchymal stem cells. Leuk Lymphoma. 46:1531–1544. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Wang Y, Wang M, Abarbanell AM, Weil BR, Herrmann JL, Tan J, Novotny NM, Coffey AC and Meldrum DR: MEK mediates the novel cross talk between TNFR2 and TGF-EGFR in enhancing vascular endothelial growth factor (VEGF) secretion from human mesenchymal stem cells. Surgery. 146:198–205. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Scherzed A, Hackenberg S, Froelich K, Kessler M, Koehler C, Hagen R, Radeloff A, Friehs G and Kleinsasser N: BMSC enhance the survival of paclitaxel treated squamous cell carcinoma cells in vitro. Cancer Biol Ther. 11:349–357. 2011. View Article : Google Scholar

19 

Park JI: Growth arrest signaling of the Raf/MEK/ERK pathway in cancer. Front Biol (Beijing). 9:95–103. 2014. View Article : Google Scholar

20 

Whyte J, Bergin O, Bianchi A, McNally S and Martin F: Key signalling nodes in mammary gland development and cancer. Mitogen-activated protein kinase signalling in experimental models of breast cancer progression and in mammary gland development. Breast Cancer Res. 11:2092009. View Article : Google Scholar : PubMed/NCBI

21 

Dhillon AS, Hagan S, Rath O and Kolch W: MAP kinase signalling pathways in cancer. Oncogene. 26:3279–3290. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Scherzed A, Hackenberg S, Radeloff A, Froelich K, Rak K, Hagen R and Kleinsasser N: Human mesenchymal stem cells promote cancer motility and cytokine secretion in vitro. Cells Tissues Organs. 198:327–337. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, Bae YC and Jung JS: Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 14:311–324. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Rangan SR: A new human cell line (FaDu) from a hypopharyngeal carcinoma. Cancer. 29:117–121. 1972. View Article : Google Scholar : PubMed/NCBI

25 

Zenner HP, Lehner W and Herrmann IF: Establishment of carcinoma cell lines from larynx and submandibular gland. Arch Otorhinolaryngol. 225:269–277. 1979. View Article : Google Scholar : PubMed/NCBI

26 

Witz IP: Tumor-microenvironment interactions: Dangerous liaisons. Adv Cancer Res. 100:203–229. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Zigrino P, Löffek S and Mauch C: Tumor-stroma interactions: Their role in the control of tumor cell invasion. Biochimie. 87:321–328. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Chen Z, Malhotra PS, Thomas GR, Ondrey FG, Duffey DC, Smith CW, Enamorado I, Yeh NT, Kroog GS, Rudy S, et al: Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin Cancer Res. 5:1369–1379. 1999.PubMed/NCBI

29 

Kanazawa T, Nishino H, Hasegawa M, Ohta Y, Iino Y, Ichimura K and Noda Y: Interleukin-6 directly influences proliferation and invasion potential of head and neck cancer cells. Eur Arch Otorhinolaryngol. 264:815–821. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Lederle W, Depner S, Schnur S, Obermueller E, Catone N, Just A, Fusenig NE and Mueller MM: IL-6 promotes malignant growth of skin SCCs by regulating a network of autocrine and paracrine cytokines. Int J Cancer. 128:2803–2814. 2011. View Article : Google Scholar

31 

Cohen S, Bruchim I, Graiver D, Evron Z, Oron-Karni V, Pasmanik-Chor M, Eitan R, Bernheim J, Levavi H, Fishman A, et al: Platinum-resistance in ovarian cancer cells is mediated by IL-6 secretion via the increased expression of its target cIAP-2. J Mol Med Berl. 91:357–368. 2013. View Article : Google Scholar

32 

Chang H, Shi Y, Tuokan T, Chen R and Wang X: Expression of aquaporin 8 and phosphorylation of Erk1/2 in cervical epithelial carcinogenesis: Correlation with clinicopathological parameters. Int J Clin Exp Pathol. 7:3928–3937. 2014.PubMed/NCBI

33 

Hsu YL, Hou MF, Kuo PL, Huang YF and Tsai EM: Breast tumor-associated osteoblast-derived CXCL5 increases cancer progression by ERK/MSK1/Elk-1/snail signaling pathway. Oncogene. 32:4436–4447. 2013. View Article : Google Scholar

34 

Shi Y, Hsu JH, Hu L, Gera J and Lichtenstein A: Signal pathways involved in activation of p70S6K and phosphorylation of 4E-BP1 following exposure of multiple myeloma tumor cells to interleukin-6. J Biol Chem. 277:15712–15720. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Levy DE and Darnell JE Jr: Stats: Transcriptional control and biological impact. Nat Rev Mol Cell Biol. 3:651–662. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Rose-John S, Scheller J, Elson G and Jones SA: Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: Role in inflammation and cancer. J Leukoc Biol. 80:227–236. 2006. View Article : Google Scholar : PubMed/NCBI

37 

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

38 

Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, Li J, Yan X, Liu Y, Shao C, et al: Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 23:925–933. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Liu R, Wei S, Chen J and Xu S: Mesenchymal stem cells in lung cancer tumor microenvironment: Their biological properties, influence on tumor growth and therapeutic implications. Cancer Lett. 353:145–152. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Direkze NC, Hodivala-Dilke K, Jeffery R, Hunt T, Poulsom R, Oukrif D, Alison MR and Wright NA: Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 64:8492–8495. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, Ganesan S, Glod JW and Banerjee D: Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 68:4331–4339. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Silzle T, Kreutz M, Dobler MA, Brockhoff G, Knuechel R and Kunz-Schughart LA: Tumor-associated fibroblasts recruit blood monocytes into tumor tissue. Eur J Immunol. 33:1311–1320. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Koukourakis MI, Giatromanolaki A, Harris AL and Sivridis E: Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: A metabolic survival role for tumor-associated stroma. Cancer Res. 66:632–637. 2006. View Article : Google Scholar : PubMed/NCBI

44 

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

45 

Qiao L, Xu ZL, Zhao TJ, Ye LH and Zhang XD: Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Lett. 269:67–77. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 47 Issue 1

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
Scherzad A, Steber M, Gehrke T, Rak K, Froelich K, Schendzielorz P, Hagen R, Kleinsasser N and Hackenberg S: Human mesenchymal stem cells enhance cancer cell proliferation via IL-6 secretion and activation of ERK1/2. Int J Oncol 47: 391-397, 2015
APA
Scherzad, A., Steber, M., Gehrke, T., Rak, K., Froelich, K., Schendzielorz, P. ... Hackenberg, S. (2015). Human mesenchymal stem cells enhance cancer cell proliferation via IL-6 secretion and activation of ERK1/2. International Journal of Oncology, 47, 391-397. https://doi.org/10.3892/ijo.2015.3009
MLA
Scherzad, A., Steber, M., Gehrke, T., Rak, K., Froelich, K., Schendzielorz, P., Hagen, R., Kleinsasser, N., Hackenberg, S."Human mesenchymal stem cells enhance cancer cell proliferation via IL-6 secretion and activation of ERK1/2". International Journal of Oncology 47.1 (2015): 391-397.
Chicago
Scherzad, A., Steber, M., Gehrke, T., Rak, K., Froelich, K., Schendzielorz, P., Hagen, R., Kleinsasser, N., Hackenberg, S."Human mesenchymal stem cells enhance cancer cell proliferation via IL-6 secretion and activation of ERK1/2". International Journal of Oncology 47, no. 1 (2015): 391-397. https://doi.org/10.3892/ijo.2015.3009