Skip to main content
Tissue Engineering and Regenerative Medicine logoLink to Tissue Engineering and Regenerative Medicine
. 2019 Nov 1;16(6):615–630. doi: 10.1007/s13770-019-00207-w

Cancer Conditioned Medium Modulates Functional and Phenotypic Properties of Human Decidua Parietalis Mesenchymal Stem/Stromal Cells

E Bahattab 1, T Khatlani 2, F M Abomaray 3,, S A Messaoudi 4, M H Abumaree 2,5,
PMCID: PMC6879693  PMID: 31824824

Abstract

Background:

Mesenchymal Stem/Stromal Cells (MSCs) from the decidua parietalis (DPMSCs) of human term placenta express several molecules with important biological and immunological properties. DPMSCs induce natural killer cell expression of inflammatory receptors and their cytotoxic activity against cancer cells. These properties make DPMSCs promising therapeutical agent for cancer. The successful development of MSCs as an anti-cancer therapeutic cells rely on their ability to function in a hostile inflammatory and oxidative stress cancer environment. Here, we studied the effects of conditioned medium obtained from the culture of breast cancer cells (CMMDA-231) on the functional and phenotypic properties of DPMSCs.

Methods:

DPMSCs were cultured with CMMDA-231 and important functions of DPMSCs were measured. The effect of CMMDA-231 on DPMSC expression of several genes with different functions was also evaluated.

Results:

DPMSCs were able to function in response to CMMDA-231, but with reduced proliferative and adhesive potentials. Preconditioning of DPMSCs with CMMDA-231 enhanced their adhesion while reducing their invasion. In addition, CMMDA-231 modulated DPMSC expression of many genes with various functional (i.e., proliferation, adhesion, and invasion) properties. DPMSCs also showed increased expression of genes with anti-cancer property.

Conclusion:

These data show the ability of DPMSCs to survive and function in cancer environment. In addition, preconditioning of DPMSCs with CMMDA-231 enhanced their anti-cancer properties and thus demonstrating their potential as an anti-cancer therapeutic agent. However, future studies are essential to reveal the mechanism underlying the effects of MDA-231 on DPMSC functional activities and also to confirm the anti-cancer therapeutic potential of DPMSCs.

Keywords: Proliferation, Adhesion, Migration, Invasion, Gene expression

Introduction

Previously, we have reported the isolation and characterization of mesenchymal stem/stromal cells (MSCs) from the maternal tissue of human term placenta, specifically from a region known as decidua parietalis(DPMSCs) [1]. These MSCs are adult multipotent cells with multipotent differentiation potential into the three mesenchymal lineages of adipocytes, osteocytes, and chondrocytes [1]. DPMSCs express several molecules with important biological and immunological properties including proliferation, adhesion, migration, invasion, angiogenesis, differentiation, immunomodulation and angiogenesis [1].

Recently, we studied the consequences of the interaction between DPMSCs and natural killer cells (NK cells), lymphocytes with specific cytotoxic immune functions against virally infected cells and tumor cells [2]. We reported that DPMSCs induce NK cell expression of inflammatory receptors [i.e., IL18 receptors (IL-18Rα, IL-18Rβ)] and Toll like receptor (TLR-7) [3]. These receptors are known for their anti-cancer functional activities. We also reported the ability of DPMSCs to enhance NK cell anti-cancer activity [3]. These results show the anti-cancer therapeutic potential of DPMSCs.

The use of MSCs as a therapy for cancers relies on the ability of transplanted MSCs to function normally in tumor microenvironment. This microenvironment shares similarity with an inflamed tissue that contains non-cellular and cellular components as well as secreted inflammatory and oxidative molecules [4]. This is why tumors are considered as a wound healing, which is defined as a dynamic process of chronic inflammation that promotes proliferation of cells and remodelling of tissues [5]. Similarly, chronic inflammation is responsible for cancer initiation (tumorigenesis) and progression (tumor metastasis) [5].

Tumor microenvironment contains proinflammatory mediators (i.e., cytokines, chemokines, and growth factors, etc.) secreted by cancer cells, cancer associated fibroblasts, tumor infiltrating immune cells and other cell types [4, 5]. This tumor microenvironment stimulates tumor growth and cancer progression. In addition, it provides a chemotactic stimuli to attract the transplanted MSCs to the tumor site where they must perform their functional activities in a harsh and hostile microenvironment [611]. Several studies reported the harmful effect of tumor microenvironment on the function of transplanted MSCs, and therefore limiting their clinical applications [612].

Therefore, for successful use of DPMSCs as therapeutic agent in cancer patients, it is necessary to investigate their functional responses to cancer environment. In this study, we examined the effects of cancer environment on the functions of DPMSCs and their expression of various genes with various functional properties. We exposed DPMSCs to CMMDA-231 [conditioned medium (CM) obtained from the culture of MDA-231 (breast cancer cell line)] and their functional and phenotypic properties were evaluated. We found that DPMSCs maintained their functional activities (i.e., proliferation, adhesion, migration and invasion) under a continuous exposure to CMMDA-231, but with reduced proliferation and adhesion while showing enhanced invasion. Preconditioning of DPMSCs by CMMDA-231 reversed the inhibitory and stimulatory effects of CMMDA-231 on DPMSC adhesion and invasion, respectively. In addition, preconditioning with CMMDA-231 modulated DPMSC expression of genes with multiple functions, such as proliferation, adhesion and invasion. Moreover, preconditioned DPMSCs showed high expression of genes with anti-cancer properties. These data show the ability of DPMSCs to function normally in cancer environment while their preconditioning by this environment increases their anti-cancer therapeutic potential.

Materials and methods

Ethical approval and the collection of tissues (human placentae and umbilical cord tissues)

The institutional review board at KAIMRC (King Abdulla International Medical Research Centre) approved this study (IRBC/1187/17). Human placentae and umbilical cord tissues were obtained from uncomplicated pregnancies (38–40 weeks of gestation) of consented donors. Tissues were processed immediately. The clinical and experimental procedures were performed as per the guidelines and regulations of KAIMRC.

Isolation and culture of DPMSCs

DPMSCs were isolated and cultured in complete DPMSC culture medium [DMEM-F12 medium containing 10% FBS (fetal bovine serum, Life Technologies), 100 μg/mL of L-glutamate, and antibiotics (100 µg/mL streptomycin and 100 U/mL penicillin)] using our previously described method [1]. DPMSCs were incubated at 37 °C in a humidified chamber consisting of 5% CO2 and 95% air, in a cell culture incubator. DPMSCs (passage 3) from a total of ten placentae were used.

Isolation and culture of HUVEC (human umbilical vein endothelial cells)

HUVEC were isolated and cultured in complete endothelial cell growth medium (Catalogue number PCS-100-041™, ATCC, Manassas, VA, USA) at 37 °C in a cell culture incubator using our previously published method [13]. HUVEC (passages 3–5) from ten umbilical cord tissues were used.

Cells (DPMSCs and HUVEC) were harvested using TrypLE™ solution (Life Technologies, Carlsbad, CA, USA), their viability was determined by Trypan blue, and then used in subsequent experiments.

DPMSC adhesion and proliferation using the xCELLigence system

Four DPMSC treatment groups were used in this study (Table 1 (i), Fig. 1). To produce CMMDA-231 [Conditioned medium harvested from the culture of breast cancer cell line, catalogue number MDA-MB-231 (ATCC® HTB-26™), ATCC, Manassas, VA, USA] supernatant from MDA-231, culture was prepared as previously described [14]. Briefly, 1 × 105 MDA-231 were cultured in a culture medium of DMEM-F12 containing 10% FBS, 100 μg/mL of L-glutamate, and antibiotics (above). Culture medium was changed every 48 h until 75% of cell confluency was achieved. Cells were fed with fresh culture medium for 72 h, and conditioned medium (CMMDA-231) was then collected by centrifugation.

Table 1.

DPMSC treatment groups used in this study

Group Description
(i) DPMSC treatment groups used in the proliferation and adhesion experiments
1 DPMSC cultured alone
2 DPMSCs cultured with 1–25% CMMDA-231
3 DPMSCs cultured with 25% CMMDA-231 [25 (in)]
4 DPMSCs precultured with 25% CMMDA-231 for 72 h [pre (25)], and DPMSCs were then harvested and used
(ii) DPMSC treatment groups used in the migration experiments
1 DPMSC cultured alone
2 DPMSCs cultured with with 25% CMMDA-231 [25 (in)] in the upper chamber
3 DPMSCs precultured with 25% CMMDA-231 for 72 h [pre (25)], and DPMSCs were then harvested and seeded in the upper chamber
(iii) DPMSC treatment groups used in the invasion experiments
1 DPMSCs were added to the monolayer of endothelial cells
2 DPMSCs were added to the monolayer of endothelial cells in the presence of 25% CMMDA-231 [25 (in)]
3 DPMSCs precultured with 25% CMMDA-231 for 72 h [pre (25)], and DPMSCs were then harvested and added to the monolayer of endothelial cells

CMMDA-231: conditioned medium experiment

Fig. 1.

Fig. 1

AD DPMSC culture system used in this study. DPMSCs were seeded on a surface of 16-well plate in the xCELLigence system containing complete DPMSC culture medium alone (Untreated DPMSC), or with 1–25% CMMDA-231 [conditioned medium (CM) obtained from the culture of MDA-231], or with 25% CMMDA-231 [25 (in)], and pre (25) [DPMSCs precultured with 25% CMMDA-231 for 72 h], and DPMSCs were then harvested and seeded in the well of 16-well plate in xCELLigence. In all culture systems, cells were incubated at 37 °C in a cell culture incubator. EG DPMSC proliferation in response to various concentrations (0–25%) of CMMDA-231 using the xCELLigence system. At 24 h, and as compared to untreated DPMSCs, DPMSC proliferation was not significantly changed in response to 1, 5 and 10% CMMDA-231 (p > 0.05), but significantly reduced in response to 25% CMMDA-231. In contrast, at 48 h, the proliferation of DPMSCs significantly reduced to 1, 5 and 25% CMMDA-231, but not to 10% CMMDA-231 (p > 0.05) as compared to untreated DPMSCs. At 72 h, DPMSC proliferation significantly reduced in response to 1 and 25% CMMDA-231, but not to 5 and 10% CMMDA-231 as compared to untreated DPMSCs, p > 0.05. HJ Reversibility of DPMSC proliferation. DPMSCs were initially cultured with 25% CMMDA-231 [25 (pre)] for 72 h, DPMSCs- pretreated with CMMDA-231 were then harvested and seeded on a surface of 16- well plate in a complete DPMSC culture medium, and their proliferation was then compared to untreated DPMSCs or DPMSCs cultured in a complete DPMSC culture medium containing 25% CMMDA-231 [25 (in)] using the xCELLigence system. At 24, 48 and 72 h, and as compared to untreated DPMSCs, the proliferation of DPMSCs cultured with 25% CMMDA-231 [25 (in)] significantly reduced. As compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)], the pretreatment of DPMSCs with 25% CMMDA-231 [25 (pre)] did not significantly change (p > 0.05) the proliferation of DPMSCs at 24 h, but it was significantly reduced at 48 and 72 h. Each experiment was performed in triplicate and repeated with five independent DPMSC (passage 3) preparations. *p < 0.05. Bars represent standard errors

The adhesion and proliferation of DPMSCs were evaluated using the xCELLigence system (RTCA-DP; Roche Diagnostics, Mannheim, Germany) as previously described [13, 1517]. Briefly, 5 × 103 DPMSCs (Table 1 (i), Fig. 1A–D) were seeded in a well of 16-well culture plates (Catalogue number 05469813001, E-Plate 16, Roche Diagnostics, Basel, Switzerland) containing complete DPMSC culture medium (Table 1 (i), Fig. 1). The culture plates were then placed in the xCELLigence system (RTCA-DP; Roche Diagnostics, Mannheim, Germany) at 37 °C in a cell culture incubator, and the cell index was automatically monitored. Data for cell adhesion (at 2 h) and proliferation (24–72 h) was measured as previously described [3, 13, 1517]. Each experiment was performed in triplicate and repeated with five independent DPMSC (passage 3) preparations.

DPMSC migration using xCELLigence system

Cell treatment groups (Table 1 (ii), Fig. 3) were used to evaluate DPMSC migration using the xCELLigence system, and CIM-16-well plates (Catalogue number 05665825001, Roche Diagnostics, Basel, Switzerland) as previously described [13, 1517]. Briefly, 1 × 104 DPMSCs were seeded in the upper chamber of the plate, and automatically monitored for 24 h by the xCELLigence system. The data was expressed as a cell index value [13, 1517]. DPMSC migration with, and without 30% FBS served as a positive and negative control, respectively. Each experiment was performed and repeated as described above.

Fig. 3.

Fig. 3

AC DPMSC migration experiments used in this study. DPMSCs cultured alone in the upper chamber, or DPMSCs cultured with 25% CMMDA-231 [25 (in)] in the upper chamber, or pre (25) [DPMSCs precultured with 25% CMMDA-231 for 72 h], and DPMSCs were then harvested and seeded in the well of 16- well plate in xCELLigence in the upper chamber. DPMSCs were seeded in DPMSC serum free medium in the upper chamber of CIM migration plate while DPMSC culture medium containing 30% FBS was added to the lower chambers. D DPMSC migration in response to CMMDA-231 or after removing the effects of CMMDA-231. DPMSCs were initially cultured with CMMDA-231 for 72 h and then cultured alone in a migration assay using the xCELLigence system. At 24 h, the migration of DPMSCs cultured with 25% CMMDA-231 [25 (in)] or pretreated with 25% CMMDA-231 [25 (pre)] did not significantly change (p > 0.05) as compared to untreated DBMSCs. The pretreatment of DPMSCs with 25% CMMDA-231 [25 (pre)] did not change the migration of DPMSCs at 24 h as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)]. Each experiment was performed in triplicate and repeated with five independent DPMSC (passage 3) preparations. *p < 0.05. Bars represent standard errors

DPMSC invasion under the effect of CMMDA-231

Using the xCELLigence system, we evaluated the effect of CMMDA-231 on the ability of DPMSCs to invade through a monolayer of endothelial cells. Briefly, 2.5 × 104 HUVEC were seeded in complete endothelial cell growth medium in a 16-well culture E-Plate (as described above) until they reached a growth plateau (20 h). Different treatments of 1 × 104 DPMSCs (Table 1 (iii)) were then added to this monolayer of endothelial cells. After 10 h, data for the invasion was recorded and expressed as cell index (mean ± standard error). The rate of cell invasion was determined by calculating the normalized cell index at pausing time (20 h) of HUVEC growth.

Gene expression by real-time polymerase chain reaction (RT-PCR)

DPMSC expression of 84 genes relating to Human Cytokines and Chemokines (Catalogue number PAHS-150Z, Qiagen, Hilden, Germany) was identified using the RT-PCR system [15, 1719]. Total RNA was extracted from DPMSCs [untreated DPMSCs, DPMSCs pretreated with 25% CMMDA-231 for 72 h, and DPMSCs pretreated with 25% CMMDA-231 for 72 h and then re-cultured alone for 72 h] and cDNA was synthesized and used in a QuantiTect Primer Assay (Qiagen, Hilden, Germany). The real-time polymerase chain reaction (RT-PCR) was performed in triplicate on the CFX96 real-time PCR detection system (BIO-RAD, Hercules, CA, USA), and the data was analyzed by calculating ΔΔ−2 values. The results are expressed as fold change expression as compared to control Relative expression of house-keeping genes were used as internal controls. Experiments were performed in triplicate and repeated three times using DPMSCs prepared from three independent placentae.

Statistical analysis

GraphPad Prism 5 was used to analyze data using non-parametric tests (Mann–Whitney U and Kruskal–Wallis). Data were deemed statistically significant if p < 0.05.

Results

CMMDA-231 reduced DPMSC proliferation

DPMSCs were used to examine the effect of CMMDA-231 on DPMSC proliferation by the xCELLigence system. At 24 h, and as compared to untreated DPMSCs, DPMSC proliferation did not significantly change in response to 1, 5, and 10% CMMDA-231 (p > 0.05), but significantly reduced (p < 0.05) in response to 25% CMMDA-231 (Fig. 1E). In contrast, at 48 h, the proliferation of DPMSCs significantly reduced (p < 0.05) in response to 1, 5 and 25% CMMDA-231, but not to 10% CMMDA-231 as compared to untreated DPMSCs (Fig. 1F). At 72 h, DPMSC proliferation significantly reduced (p < 0.05) in response to 1 and 25% CMMDA-231, but not to 5 and 10% CMMDA-231 (p > 0.05) as compared to untreated DPMSCs (Fig. 1G).

The viability of DPMSCs treated with CMMDA-231 (1–25%) for all examined time points was 95%. In some experiments, DPMSCs were cultured with 50 and 100% CMMDA-231, and their viability at these two concentrations was > 90%. Based on the results obtained above, the exposure time of 72 h and CMMDA-231 at 25% were selected to evaluate the effect of CMMDA-231 on DPMSC functions (proliferation, adhesion, migration, and invasion).

Reversibility of DPMSC proliferation under the effect of CMMDA-231

To evaluate the reversibility of the inhibitory effect of CMMDA-231 on DPMSC proliferation, cells were intially cultured with 25% CMMDA-231 [25 (pre)] for 72 h, harvested, and then their proliferation as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)] was determined using the xCELLigence system.

At 24, 48 and 72 h, and as compared to untreated DPMSCs, the proliferation of DPMSCs cultured with 25% CMMDA-231 [1 (in)] or precultured with 25% CMMDA-231 for 72 h [25 (pre)] significantly reduced (p < 0.05) while the pretreatment of DPMSCs with 25% CMMDA-231 for 72 h [25 (pre)] had no significant effect (p > 0.05) on their proliferation as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)] at 24 h, but was significantly reduced (p < 0.05) at 48 and 72 h (Fig. 1H–J).

CMMDA-231 effect on DPMSC adhesion

To evaluate the effect of CMMDA-231 on DPMSC adhesion, the xCELLigence system was used. At 2 h, and as compared to untreated DPMSCs, the adhesion of DPMSCs cultured with 25% CMMDA-231 [1 (in)], but not precultured with 25% CMMDA-231 for 72 h [25 (pre)] significantly reduced (p < 0.05). In addition, the pretreatment of DPMSCs with 25% CMMDA-231 for 72 h [25 (pre)] significantly increased (p < 0.05) DPMSC adhesion as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)] at 2 h (Fig. 2).

Fig. 2.

Fig. 2

DPMSC adhesion in response to CMMDA-231 or after removing the effects of CMMDA-231. DPMSCs were initially cultured with CMMDA-231 for 72 h and then cultured alone in an adhesion assay using the xCELLigence system. At 2 h, and as compared to untreated DPMSCs, the adhesion of DPMSCs significantly reduced in response to 25% CMMDA-231 [25 (in)]. The pretreatment of DPMSCs with 25% CMMDA-231 [25 (pre)] did not significantly change (p > 0.05) the adhesion of DPMSCs at 2 h as compared to untreated DPMSCs, but as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)], the DPMSC adhesion significantly increased. Each experiment was performed in triplicate and repeated with five independent DPMSC (passage 3) preparations. *p < 0.05. Bars represent standard errors

CMMDA-231 effect on DPMSC migration

We also evaluated the effect of CMMDA-231 on the migration of DPMSCs cultured alone or with 25% CMMDA-231 [25 (in)] or precultured with 25% CMMDA-231 [25 (pre)] in the upper chamber of the plate using the xCELLigence system. At 24 h, the migration of DPMSCs cultured with 25% CMMDA-231 [25 (in)] or precultured with 25% CMMDA-231 [25 (pre)] did not significantly change (p > 0.05) as compared to untreated DPMSCs (Fig. 3). The pre-culture of DPMSCs with 25% CMMDA-231 [25 (pre)] did not significantly change (p > 0.05) the migration of DPMSCs at 24 h and as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)] (Fig. 3D).

CMMDA-231 effect on DPMSC invasion of HUVEC

We also evaluated the effect of CMMDA-231 on DPMSC invasion of HUVEC by the xCELLigence system. Invasion is defined as the infiltration of HUVEC monolayer by DPMSCs which causes detachment of HUVEC that results in increased adhesion of DPMSCs associated with elevation in the cell index. At 10 h treatment and as compared to untreated DPMSCs, the invasion of HUVEC monolayer by DPMSCs cultured in 25% CMMDA-231 was significantly increased (p < 0.05) (Fig. 4), but the invasion of HUVEC monolayer by DPMSCs pretreated with 25% CMMDA-231 for 72 h [25 (pre)] did not significantly change (p > 0.05) as compared to DPMSCs cultured in 25% CMMDA-231 [25 (in)] (Fig. 4). In addition, pretreatment of DPMSCs with 25% CMDA-231 for 72 h [25 (pre)] significantly reduced (p < 0.05) DPMSC invasion of HUVEC monolayer as compared to DPMSCs cultured in 25% CMMDA-231 [25 (in)] (Fig. 4).

Fig. 4.

Fig. 4

Effects of CMMDA-231 on DPMSC invasion through HUVEC monolayer. DPMSC invasion was examined by adding DPMSCs to a monolayer of HUVEC, and the invasion of DPMSCs through HUVEC monolayer was then assessed by the xCELLigence system. Invasion is defined as infiltration of HUVEC monolayer by DPMSCs which causes detachment of HUVEC resulting in increased adhesion of DPMSCs leading to elevated cell index. At 10 h and as compared to untreated DPMSCs, the invasion of DPMSCs cultured with 25% CMMDA-231 [25 (in)] significantly increased. The pretreatment of DPMSCs with 25% CMMDA-231 did not significantly change (p > 0.05) the invasion of DPMSCs as compared to untreated DPMSCs, but as compared to DPMSCs cultured with 25% CMMDA-231 [25 (in)], the invasion of DPMSCs significantly reduced. Each experiment was performed in triplicate and repeated with five independent DPMSC (passage 3) preparations. *p < 0.05. Bars represent standard errors

CMMDA-231 modulated the expression of genes important in DPMSC functions

The RT-PCR results show that 25% CMMDA-231 modulated the expression of many genes in DPMSCs (Tables 2, 3, 4, 5, 6).

Table 2.

MDA-231 modulated DPMSC expression of genes with pro-proliferative and anti-proliferative properties

No. Gene symbol Gene full name CMDPMSC mean ΔΔ−2 values Pre-CMDPMSC mean ΔΔ−2 values Fold change (Pre-CMDPMSC vs. CMDPMSC) p < 0.05 Biological activities
1 CCL2 C-C motif chemokine ligand 2 239.78 2.10 114.18 Fold Promotes proliferation [36]
2 CCL18 C-C motif chemokine ligand 18 9.33 2.27 4.11 Fold Promotes proliferation [37]
3 CCL19 C-C motif chemokine ligand 19 474.38 8.69 54.58 Fold Promote proliferation
4 CCL20 C-C motif chemokine ligand 20 432.53 2.30 188.05 Fold Promotes proliferation [38]
5 IL-4 Interleukin-4 711.43 24.60 28.91 Fold Promotes proliferation [39]
6 IL-6 Interleukin-6 2.96 12.02 4.06 Fold Promotes proliferation [40]
7 IL-7 Interleukin-7 1.92 0.37 5.18 Fold Promotes proliferation [41]
8 IL-16 Interleukin-16 87.96 1.37 64.20 Fold Promotes proliferation [42]
9 CSF2 Colony stimulating factor 2 4.05 1.31 3.09 Fold Promotes proliferation [43]
10 LTA Lymphotoxin alpha 19.17 3.23 5.93 Fold Promotes proliferation [44]
11 THPO Thrombopoietin 4.33 1.49 2.90 Fold Promotes proliferation [45]
12 TNF Tumor necrosis factor 2.85 1.40 2.03 Fold Promotes proliferation [46]
13 VEGFA Vascular endothelial growth factor A 2.92 1.10 2.65 Fold Promotes proliferation [47]
14 CCL13 C-C motif chemokine ligand 13 0.86 2.78 3.23 Fold Promotes proliferation [95]
15 CCL24 C-C motif chemokine ligand 24 0.50 2.35 4.70 Fold Promotes proliferation [96]
16 CXCL16 C-X-C motif chemokine ligand 16 0.50 86.88 173.76 Fold Promotes proliferation [97]
17 CNTF Ciliary neurotrophic factor 1.25 2.41 1.92 Fold Promotes proliferation [98]
18 CSF1 Colony stimulating factor 1 0.32 1.67 5.21 Fold Promotes proliferation [85]
19 BMP2 Bone morphogenetic protein 2 0.90 1.65 1.83 Fold Promotes proliferation [99]
20 GBI Glucose-6-phosphate isomerase 0.55 2.80 5.09 Fold Promote proliferation [100]
21 IL-1RN Interleukin-1 receptor antagonist 0.69 1.83 2.65 Fold Inhibits proliferation [48]
22 IL-11 Interleukin-11 29.50 12.60 2.34 Fold Inhibits proliferation [49]
23 IL-24 Interleukin-24 3.60 1.46 2.46 Fold Inhibits proliferation [50]
24 IFNγ Interferon gamma 0.0003 4.96 16,533 Fold Inhibits proliferation [51]

CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h). Pre-CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h, and were then re-cultured alone for another 72 h)

Table 3.

MDA-231 modulated DPMSC expression of genes with adhesive, migratory and invasive properties

No. Gene symbol Gene full name CMDPMSC mean ΔΔ−2 values Pre-CMDPMSC mean ΔΔ−2 values Fold change (Pre-CMDPMSC vs. CMDPMSC) p < 0.05 Biological activities
1 BMP2 Bone morphogenetic protein 2 0.90 1.65 1.83 Fold Promotes adhesion [55]
2 CX3CL1 Chemokine (C-X3-C motif) ligand 1 0.000079 0.0099 125.31 Fold Promotes adhesion [56]
3 VEGFA Vascular endothelial growth factor A 2.92 1.10 2.65 Fold Promotes adhesion [101]
4 CCL3 C-C motif chemokine ligand 3 5.74 15.94 2.77 Fold Promote migration [65]
5 CCL18 C-C motif chemokine ligand 18 9.33 2.27 4.11 Fold Promotes migration [57]
6 CXCL12 C-X-C motif chemokine ligand 12 7.41 1.80 4.11 Fold Promotes migration [58]
7 CXCL5 C-X-C motif chemokine ligand 5 4.78 40.89 8.55 Fold Promotes migration [59]
8 IL-4 Interleukin-4 711.43 24.60 28.91 Fold Promotes migration [60]
9 IL-11 Interleukin-11 29.50 12.60 2.34 Fold Promotes migration [49]
10 IL-16 Interleukin-16 87.96 1.37 64.20 Fold Promotes migration [61]
11 IL-24 Interleukin-24 3.60 1.46 2.46 Fold ↓  Inhibits migration [62]
12 TNF Tumor necrosis factor 2.85 1.40 2.03 Fold Promotes migration [63]
13 VEGFA vascular endothelial growth factor A 2.92 1.10 2.65 Fold Promotes migration [47]
14 BMP2 Bone morphogenetic protein 2 0.90 1.65 1.83 Fold Promotes migration [64]
15 CCL2 C-C motif chemokine ligand 2 239.78 2.10 114.18 Fold Promotes invasion [66]
16 CCL18 C-C motif chemokine ligand 18 9.33 2.27 4.11 Fold Promotes invasion [37]
17 CCL20 C-C motif chemokine ligand 20 432.53 2.30 188.05 Fold Promotes invasion [38]
18 CXCL12 C-X-C motif chemokine ligand 12 7.41 1.80 4.11 Fold Promotes invasion [58]
19 IL-6 Interleukin-6 2.96 12.02 4.06 Fold Promotes invasion [67]
20 IL-11 Interleukin-11 29.50 12.60 2.34 Fold Inhibits invasion [69]
21 VEGFA Vascular endothelial growth factor A 2.92 1.10 2.65 Fold Promotes invasion [68]
22 CCL13 C-C motif chemokine ligand 13 0.86 2.78 3.23 Fold Promotes invasion [95]
23 CCL24 C-C motif chemokine ligand 24 0.50 2.35 4.70 Fold Promotes invasion [96]
24 CXCL5 C-X-C motif chemokine ligand 5 4.78 40.89 8.55 Fold Promotes invasion [59]
25 CXCL16 C-X-C motif chemokine ligand 16 0.50 86.88 173.76 Fold Promotes invasion [97]
26 IL-24 Interleukin-24 3.60 1.46 2.46 Fold Inhibits invasion [62]
27 BMP2 Bone morphogenetic protein 2 0.90 1.65 1.83 Fold Promotes invasion [99]

CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h). Pre-CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h, and were then re-cultured alone for another 72 h)

Table 4.

MDA-231 modulated DPMSC expression of genes with inflammatory properties

No. Gene symbol Gene full name CMDPMSC mean ΔΔ−2 values Pre-CMDPMSC mean ΔΔ−2 values Fold change (Pre-CMDPMSC vs. CMDPMSC) p < 0.05 Biological activities
1 CCL3 C-C motif chemokine ligand 3 5.74 15.94 2.77 Fold Promotes inflammation [70]
2 CCL13 C-C motif chemokine ligand 13 0.86 2.78 3.23 Fold Promotes inflammation [71]
3 CXCL9 C-X-C motif chemokine ligand 9 2.49 19.14 7.68 Fold Promotes inflammation [72]
4 CX3CL1 Chemokine (C-X3-C motif) ligand 1 0.000079 0.0099 125.31 Fold Promotes inflammation [56]
5 IL-12A Interleukin-12A 0.64 6.10 9.53 Fold Promotes inflammation [73]
6 IL-18 Interleukin-18 1.31 2.29 1.74 Fold Promotes inflammation [74]
7 BMP2 Bone morphogenetic protein 2 0.90 1.65 1.83 Fold Promotes inflammation [75]
8 IL-11 Interleukin-11 29.50 12.60 2.34 Fold Inhibits inflammation [76]
9 CCL2 C-C motif chemokine ligand 2 239.78 2.10 114.18 Fold Promotes inflammation [77
10 CCL19 C-C motif chemokine ligand 19 474.38 8.69 54.58 Fold Promotes inflammation [78]
11 CXCL2 C-X-C motif chemokine ligand 2 1576997 9071 173.85 Fold Promotes inflammation [79]
12 IL-7 Interleukin-7 1.92 0.37 5.18 Fold Promotes inflammation [80]
13 IL-12B Interleukin-12B 19386 12.80 1514.53 Promotes inflammation [73]
14 IL-16 Interleukin-16 87.96 1.37 64.20 Fold Promotes inflammation [81]
15 LTA Lymphotoxin alpha 19.17 3.23 5.93 Fold Promotes inflammation [82
16 TNF Tumor necrosis factor 2.85 1.40 2.03 Fold Promotes and inhibits inflammation [83]
17 CNTF Ciliary neurotrophic factor 1.25 2.41 1.92 Fold Inhibits inflammation [84]
18 CSF1 Colony stimulating factor 1 0.32 1.67 5.21 Fold Inhibits inflammation [85]
19 TGFB2 Transforming growth factor-β-2 0.25 1.69 6.76 Fold Inhibits inflammation [86

CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h). Pre-CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h, and were then re-cultured alone for another 72 h)

Table 5.

MDA-231 modulated DPMSC expression of genes with protumorigenic and antitumor activities

No. Gene symbol Gene full name CMDPMSC mean ΔΔ−2 values Pre-CMDPMSC mean ΔΔ−2 values Fold change (Pre-CMDPMSC vs. CMDPMSC)
p < 0.05
Biological activities
1 CCL3 C-C motif chemokine ligand 3 5.74 15.94 2.77 Fold Antitumor activity [87]
2 IFNA2 Interferon alpha 2 31.61 4.34 7.28 Fold Antitumor activity [88]
3 IFNγ Interferon gamma 0.0003 4.96 16,533 Fold Antitumor Activity [51]
4 IL1RN Interleukin-1 receptor antagonist 0.69 1.83 2.65 Fold Antitumor activity [48]
5 IL-24 Interleukin-24 3.60 1.46 2.46 Fold Antitumor activity [50]
6 IL-4 Interleukin-4 711.43 24.60 28.91 Fold Antitumor activity [89]
7 LTA Lymphotoxin alpha 19.17 3.23 5.93 Fold Antitumor activity [90]
8 FASLG Fas ligand 1.38 0.77 1.79 Fold Antitumor activity [91]
9 IL-6 Interleukin-6 2.96 12.02 4.06 Fold Protumorigenic activity [92]
10 IL-7 Interleukin-7 1.92 0.37 5.18 Fold Protumorigenic activity [93]
11 IL16 Interleukin 16 87.96 1.37 64.20 Fold Protumorigenic activity [94]
12 TNF Tumor necrosis factor 2.85 1.40 2.03 Fold Protumorigenic activity [83]
13 VEGFA Vascular endothelial growth factor A 2.92 1.10 2.65 Protumorigenic activity [47]

CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h). Pre-CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h, and were then re-cultured alone for another 72 h)

Table 6.

MDA-231 modulated the expression of genes in DPMSCs

No. Gene symbol CMDPMSC mean ΔΔ−2 values Pre-CMDPMSC mean ΔΔ−2 values Fold change (Pre-CMDPMSC vs. CMDPMSC)
1 CCL5 0.77 1.02 Fold change is not statistically significant, p > 0.05
2 CCL7 2.30 2.46
3 CCL11 0.16 0.16
4 CCL21 0.98 1.15
5 CXCL10 0.91 0.84
6 XCL1 1.27 1.26
7 IL1A 1.05 1.45
8 IL1B 1.60 2.24
9 IL-5 0.95 1.00
10 IL-8 0.59 0.85
11 IL-10 1.36 1.09
12 IL-13 1.37 1.77
13 IL-15 1.20 0.97
14 IL-23A 1.10 1.07
15 IL-27 2.25 1.81
16 LIF 2.73 2.28
17 LTB 0.79 1.36
18 MIF 24.20 24.73
19 CD40LG 1.31 1.27
20 CSF3 1.67 2.00
21 MSTN 0.76 0.95
23 NODAL 2.40 1.13
24 OSM 1.30 1.622
25 PPBP 1.42 1.37
26 SPP1 0.88 1.32
27 TNFRSF11B 2.59 5.51
28 TNFSF10 1.49 3.04
29 TNFSF11 0.93 1.47
30 TNFSF13B 0.50 1.63
31 ADIPOQ 1.36 1.55
32 BMP4 1.03 1.31
33 BMP6 1.43 2.10
34 BMP7 1.46 1.77
35 C5 1.47 2.35

CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h). Pre-CMDPMSC (DPMSCs were cultured with 25% MDA-231 for 72 h, and were then re-cultured alone for another 72 h)

Discussion

Previously, we have reported the phenotypic and functional properties of DPMSCs [1], and their interaction with NK cells [3]. We showed that DPMSCs can induce NK cell cytotoxic activity against cancer cells [3]. This property makes DPMSCs as potential therapeutic agent for cancer. Cancer is an inflammatory disease that is characterized by a hostile microenvironment, which supports the growth and progression of tumor cells [20]. This microenvironment contains inflammatory molecules including cytokines, chemokines, growth factors that modulate the functions of transplanted MSCs [611]. Therefore, for successful use of DPMSCs as an anti-cancer therapeutic agent, we studied the effects of CMMDA-231 on DPMSC functional and phenotypic properties.

First, we report the survival of DPMSCs under a continuous exposure to high concentration (up to 100%) of CMMDA-231. MDA-231 secretes several inflammatory molecules with ability to induce cellular injury, such as IL-1β, IL-6, IL-8, and matrix metalloproteinases (MMPs) [2128]. For example, the culture of pancreatic beta-cells with IL-6 induces their apoptotic death [27].

Survival of DPMSCs in such harsh tumor environment could be attributed to their placental niche, where they are usually exposed to high levels of oxidative stress and inflammatory mediators during normal pregnancy [29, 30]. Therefore, DPMSCs might have acquired biological characteristics to resist death induced by inflammatory molecules.

The ability of DPMSCs to counteract injury induced by CMMDA-231 was further supported by maintaining their functional activities. DPMSCs proliferated in response to CMMDA-231 (Fig. 1). However, they showed a reduction in their proliferation (Fig. 1). This data may suggest that it is safe to use DPMSCs as previously reported for other types of MSCs. It was shown that the transplantation of human bone marrow derived MSCs (BMMSCs) does not form malignant tumor [31]. Therefore, the use of DPMSCs as therapeutic agent is probably safe, but requires further confirmation in animal studies before their applications in patients.

MDA-231 secretes IL-10, plasminogen activator inhibitor type 1 (PAI-1), and parathyroid hormone-related protein (PTHrP) [22, 32, 33]. These molecules have antiproliferative properties [3335]. Therefore, the antiproliferative effect of CMMDA-231 on DPMSCs is possibly mediated by these molecules. In this study, preconditioning of DPMSCs with CMMDA-231 reduced and increased their expression of pro-proliferative [3647], and antiproliferative [4851] genes, respectively, Table 2. Therefore, these genes may mediate the anti-proliferative effect of MDA-231 on DPMSCs. However, a future mechanistic study is essential to elucidate this finding further.

After transplanting MSCs into patients, MSCs must function in a disease environment where they is high level of oxidative stress and inflammatory mediators [52, 53]. In this study, CMMDA-231 reduced DPMSC adhesion. IL-10 usually reduces cell adhesion [54], and therefore, IL-10 in the conditioned medium of MDA-231, may act on DPMSCs to reduce their adhesion. Preconditioning of DPMSCs with CMMDA-231 reversed this inhibitory effect of CMMDA-231 on DPMSC adhesion (Fig. 2). This reversible effect of MDA-231 on DPMSC adhesion is probably mediated by inducing the expression of adhesive genes (i.e., BMP2 and CX3CL1) [55, 56] in preconditioned DPMSCs (Table 3). In contrast, DPMSC migration was not changed by CMMDA-231 (Fig. 3), but phenotypically, CMMDA-231 modulated DPMSC expression of genes with migratory properties [47, 49, 5765] (Table 3). However, these genes are also involved in another cellular functions, such as proliferation, adhesion, invasion, inflammation, pro-tumour and antitumor activities. Therefore these genes may mediate another functional activities of DPMSCs.

MSCs must also penetrate the endothelial cell barrier in order to reach their cellular targets. CMMDA-231 promoted DPMSC invasion on a reversible effect (Fig. 4). CMMDA-231 also modulated DPMSCs expression of invasive genes [37, 38, 58, 59, 60, 6769], Table 3. However, the molecular pathway involved in MDA-231 modulating DPMSC invasion needs to be further investigated.

Phenotypically, CMMDA-231 modulated DPMSCs expression of genes with inflammatory [57, 7076], anti-inflammatory [73, 7786], anti-tumorigenic and pro-tumorigenic properties [47, 48, 50, 51, 83, 8794] (Tables 4, 5). These phenotypic data show the double-edged sword properties of DPMSCs as we previously reported for MSCs isolated from the decidua basalis region of human term placenta [15]. Importantly, DPMSCs show anti-cancer phenotypic properties, which further confirms our previous report of DPMSCs on the anti-cancer activity of NK cells [3].

In conclusion, this study shows the functional activities of DPMSCs in response to cancer environment, and their biosafety. DPMSCs were able to perform invasion activity, and thus demonstrating their engraftment property. CMMDA-231 also modulated the phenotypic features of DPMSCs particularly their anti-cancer property. These data show the suitability of using DPMSCs in treating cancers. However, future studies will reveal the mechanism underlying the effects of MDA-231 on DPMSC functions, and confirm their anti-cancer therapeutic potential.

Acknowledgements

We appreciate the staff and patients of King Abdul Aziz Medical City for providing us with placentae. MHA proposed and supervised the project. MHA designed the experiments. EB performed the experiments. MHA, EB and TK analysed the data. MHA wrote the manuscript. MHA, FMA, TK, and SAM contributed to data analysis and interpretation of results. All authors reviewed the manuscript. This study was supported by Grants from KAIMRC (Grant No. RC12/133).

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical statement

IRB of KAIMRC approved this study (IRBC/1187/17). Samples (Placentae and umbilical cords) were obtained after signing consent forms.

Footnotes

M. H. Abumaree—deceased.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Contributor Information

F. M. Abomaray, Email: fawaz.abomaray@ki.se

M. H. Abumaree, Email: mohamedabumaree@hotmail.com, Email: abumareem@ksau-hs.edu.sa

References

  • 1.Abumaree MH, Abomaray FM, Alshehri NA, Almutairi A, AlAskar AS, Kalionis B, et al. Phenotypic and functional characterization of mesenchymal stem/multipotent stromal cells from Decidua Parietalis of human term placenta. Reprod Sci. 2016;23:1193–1207. doi: 10.1177/1933719116632924. [DOI] [PubMed] [Google Scholar]
  • 2.Brandstadter JD, Yang Y. Natural killer cell responses to viral infection. J Innate Immun. 2011;3:274–279. doi: 10.1159/000324176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Abumaree MH, Bahattab E, Alsadoun A, Al Dosaimani A, Abomaray FM, Khatlani T, et al. Characterization of the interaction between human decidua parietalis mesenchymal stem/stromal cells and natural killer cells. Stem Cell Res Ther. 2018;9:102. doi: 10.1186/s13287-018-0844-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Uchibori R, Okada T, Ito T, Urabe M, Mizukami H, Kume A, et al. Retroviral vector-producing mesenchymal stem cells for targeted suicide cancer gene therapy. J Gene Med. 2009;11(5):373–381. doi: 10.1002/jgm.1313. [DOI] [PubMed] [Google Scholar]
  • 5.Arnold KM, Opdenaker LM, Flynn D, Sims-Mourtada J. Wound healing and cancer stem cells: inflammation as a driver of treatment resistance in breast cancer. Cancer Growth Metastasis. 2015;8:1–13. doi: 10.4137/CGM.S11286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, et al. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci. 2013;3:5. doi: 10.1186/2045-3701-3-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Burr SP, Dazzi F, Garden OA. Mesenchymal stromal cells and regulatory T cells: the Yin and Yang of peripheral tolerance? Immunol Cell Biol. 2013;91:12–18. doi: 10.1038/icb.2012.60. [DOI] [PubMed] [Google Scholar]
  • 8.English K. Mechanisms of mesenchymal stromal cell immunomodulation. Immunol Cell Biol. 2013;91(1):19–26. doi: 10.1038/icb.2012.56. [DOI] [PubMed] [Google Scholar]
  • 9.Alphonso A, Alahari SK. Stromal cells and integrins: conforming to the needs of the tumor microenvironment. Neoplasia. 2009;11(12):1264–1271. doi: 10.1593/neo.91302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lee SH, Jeong D, Han YS, Baek MJ. Pivotal role of vascular endothelial growth factor pathway in tumor angiogenesis. Ann Surg Treat Res. 2015;89:1–8. doi: 10.4174/astr.2015.89.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Guan X. Cancer metastases: challenges and opportunities. Acta Pharm Sin B. 2015;5:402–418. doi: 10.1016/j.apsb.2015.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lee HY, Hong IS. Double-edged sword of mesenchymal stem cells: cancer-promoting versus therapeutic potential. Cancer Sci. 2017;108(10):1939–1946. doi: 10.1111/cas.13334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Alshabibi MA, Al Huqail AJ, Khatlani T, Abomaray FM, Alaskar AS, Alawad AO, et al. mesenchymal stem/multipotent stromal cells from human decidua basalis reduce endothelial cell activation. Stem Cells Dev. 2017;26:1355–1373. doi: 10.1089/scd.2017.0096. [DOI] [PubMed] [Google Scholar]
  • 14.Abumaree MH, Al Jumah MA, Kalionis B, Jawdat D, Al Khaldi A, Abomaray FM, et al. Human placental mesenchymal stem cells (pMSCs) play a role as immune suppressive cells by shifting macrophage differentiation from inflammatory M1 to anti-inflammatory M2 macrophages. Stem Cell Rev. 2013;9:620–641. doi: 10.1007/s12015-013-9455-2. [DOI] [PubMed] [Google Scholar]
  • 15.Alshabibi MA, Khatlani T, Abomaray FM, AlAskar AS, Kalionis B, Messaoudi SA, et al. Human decidua basalis mesenchymal stem/stromal cells protect endothelial cell functions from oxidative stress induced by hydrogen peroxide and monocytes. Stem Cell Res Ther. 2018;9(1):275. doi: 10.1186/s13287-018-1021-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Abumaree MH, Hakami M, Abomaray FM, Alshabibi MA, Kalionis B, Al Jumah MA, et al. Human chorionic villous mesenchymal stem/stromal cells modify the effects of oxidative stress on endothelial cell functions. Placenta. 2017;59:74–86. doi: 10.1016/j.placenta.2017.05.001. [DOI] [PubMed] [Google Scholar]
  • 17.Basmaeil YS, Al Subayyil AM, Khatlani T, Bahattab E, Al-Alwan M, Abomaray FM, et al. Human chorionic villous mesenchymal stem/stromal cells protect endothelial cells from injury induced by high level of glucose. Stem Cell Res Ther. 2018;9:238. doi: 10.1186/s13287-018-0984-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Abomaray FM, Al Jumah MA, Alsaad KO, Jawdat D, Al Khaldi A, AlAskar AS, et al. Phenotypic and functional characterization of mesenchymal stem/multipotent stromal cells from decidua basalis of human term placenta. Stem Cells Int. 2016;2016:5184601. doi: 10.1155/2016/5184601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Khatlani T, Algudiri D, Alenzi R, Al Subayyil AM, Abomaray FM, Bahattab E, et al. Preconditioning by hydrogen peroxide enhances multiple properties of human decidua basalis mesenchymal stem/multipotent stromal cells. Stem Cells Int. 2018;2018:6480793. doi: 10.1155/2018/6480793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gkretsi V, Stylianou A, Papageorgis P, Polydorou C, Stylianopoulos T. Remodeling components of the tumor microenvironment to enhance cancer therapy. Front Oncol. 2015;5:214. doi: 10.3389/fonc.2015.00214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Escobar P, Bouclier C, Serret J, Bièche I, Brigitte M, Caicedo A, et al. IL-1β produced by aggressive breast cancer cells is one of the factors that dictate their interactions with mesenchymal stem cells through chemokine production. Oncotarget. 2015;6:29034–29047. doi: 10.18632/oncotarget.4732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lieblein JC, Ball S, Hutzen B, Sasser AK, Lin HJ, Huang HJ, et al. STAT3 can be activated through paracrine signaling in breast epithelial cells. BMC Cancer. 2008;8:302. doi: 10.1186/1471-2407-8-302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Freund A, Chauveau C, Brouillet JP, Lucas A, Lacroix M, Licznar A, et al. IL-8 expression and its possible relationship with estrogen-receptor-negative status of breast cancer cells. Oncogene. 2003;22:256–265. doi: 10.1038/sj.onc.1206113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bachmeier BE, Nerlich AG, Lichtinghagen R, Sommerhoff CP. Matrix metalloproteinases (MMPs) in breast cancer cell lines of different tumorigenicity. Anticancer Res. 2001;21(6A):3821–3828. [PubMed] [Google Scholar]
  • 25.Shen J, Xu S, Zhou H, Liu H, Jiang W, Hao J, et al. IL-1β induces apoptosis and autophagy via mitochondria pathway in human degenerative nucleus pulposus cells. Sci Rep. 2017;7:41067. doi: 10.1038/srep41067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Thirumangalakudi L, Yin L, Rao HV, Grammas P. IL-8 induces expression of matrix metalloproteinases, cell cycle and pro-apoptotic proteins, and cell death in cultured neurons. J Alzheimers Dis. 2007;11(3):305–311. doi: 10.3233/jad-2007-11307. [DOI] [PubMed] [Google Scholar]
  • 27.Oh YS, Lee YJ, Park EY, Jun HS. Interleukin-6 treatment induces beta-cell apoptosis via STAT-3-mediated nitric oxide production. Diabetes Metab Res Rev. 2011;27:813–819. doi: 10.1002/dmrr.1233. [DOI] [PubMed] [Google Scholar]
  • 28.Tan RJ, Fattman CL, Niehouse LM, Tobolewski JM, Hanford LE, Li Q, et al. Matrix metalloproteinases promote inflammation and fibrosis in asbestos-induced lung injury in mice. Am J Respir Cell Mol Biol. 2006;35:289–297. doi: 10.1165/rcmb.2005-0471OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Raijmakers MT, Roes EM, Poston L, Steegers EA, Peters WH. The transient increase of oxidative stress during normal pregnancy is higher and persists after delivery in women with pre-eclampsia. Eur J Obstet Gynecol Reprod Biol. 2008;138(1):39–44. doi: 10.1016/j.ejogrb.2007.08.005. [DOI] [PubMed] [Google Scholar]
  • 30.Burton GJ, Jauniaux E. Oxidative stress. Best Pract Res Clin Obstet Gynaecol. 2011;25:287–299. doi: 10.1016/j.bpobgyn.2010.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bernardo ME, Zaffaroni N, Novara F, Cometa AM, Avanzini MA, Moretta A, et al. Human bone marrow derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res. 2007;67:9142–9149. doi: 10.1158/0008-5472.CAN-06-4690. [DOI] [PubMed] [Google Scholar]
  • 32.Zhang W, Xu J, Fang H, Tang L, Chen W, Sun Q, et al. Endothelial cells promote triple-negative breast cancer cell metastasis via PAI-1 and CCL5 signaling. FASEB J. 2018;32(1):276–288. doi: 10.1096/fj.201700237RR. [DOI] [PubMed] [Google Scholar]
  • 33.Morgan H, Tumber A, Hill PA. Breast cancer cells induce osteoclast formation by stimulating host IL-11 production and downregulating granulocyte/macrophage colony-stimulating factor. Int J Cancer. 2004;109:653–660. doi: 10.1002/ijc.20056. [DOI] [PubMed] [Google Scholar]
  • 34.Taga K, Tosato G. IL-10 inhibits human T cell proliferation and IL-2 production. J Immunol. 1992;148:1143–1148. [PubMed] [Google Scholar]
  • 35.Kouri FM, Queisser MA, Königshoff M, Chrobak I, Preissner KT, Seeger W, et al. Plasminogen activator inhibitor type 1 inhibits smooth muscle cell proliferation in pulmonary arterial hypertension. Int J Biochem Cell Biol. 2008;40:1872–1882. doi: 10.1016/j.biocel.2008.01.028. [DOI] [PubMed] [Google Scholar]
  • 36.Lee H, Kang JE, Lee JK, Bae JS, Jin HK. Bone-marrow-derived mesenchymal stem cells promote proliferation and neuronal differentiation of Niemann-Pick type C mouse neural stem cells by upregulation and secretion of CCL2. Hum Gene Ther. 2013;24:655–669. doi: 10.1089/hum.2013.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bo S, Donghao S, Guangqi K, Ye T. CC chemokine ligand 18 promotes cell proliferation and metastasis of urothelial carcinoma via activating PI3 K/mTOR signaling in patient with renal transplantation. Urol Int. 2018;101(4):450–458. doi: 10.1159/000492180. [DOI] [PubMed] [Google Scholar]
  • 38.Lu E, Su J, Zhou Y, Zhang C, Wang Y. CCL20/CCR39 promotes cell proliferation and metastasis in laryngeal cancer by activating p38 pathway. Biomed Pharmacother. 2017;85:486–492. doi: 10.1016/j.biopha.2016.11.055. [DOI] [PubMed] [Google Scholar]
  • 39.Or R, Renz H, Terada N, Gelfand EW. IL-4 and IL-2 promote human T-cell proliferation through symmetrical but independent pathways. Clin Immunol Immunopathol. 1992;64:210–217. doi: 10.1016/0090-1229(92)90202-y. [DOI] [PubMed] [Google Scholar]
  • 40.Li B, Jones LL, Geiger TL. IL-6 promotes T cell proliferation and expansion under inflammatory conditions in association with low-level RORγt expression. J Immunol. 2018;201:2934–2946. doi: 10.4049/jimmunol.1800016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Li WQ, Jiang Q, Aleem E, Kaldis P, Khaled AR, Durum SK. IL-7 promotes T cell proliferation through destabilization of p27Kip1. J Exp Med. 2006;203:573–582. doi: 10.1084/jem.20051520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mahindra A, Anderson KC. Role of interleukin 16 in multiple myeloma pathogenesis: a potential novel therapeutic target? J Natl Cancer Inst. 2012;104:964–965. doi: 10.1093/jnci/djs274. [DOI] [PubMed] [Google Scholar]
  • 43.Jeong W, Kim J, Bazer FW, Song G. Proliferation-stimulating effect of colony stimulating factor 2 on porcine trophectoderm cells is mediated by activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase. PLoS One. 2014;9:e88731. doi: 10.1371/journal.pone.0088731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yang JG, Wang WM, Xia HF, Yu JG, Li HM, Ren JG, et al. Lymphotoxin-alpha promotes tumor angiogenesis in HNSCC by modulating glycolysis in a PFKFB3-dependent manner. Int J Cancer. 2019;145:1358–1370. doi: 10.1002/ijc.32221. [DOI] [PubMed] [Google Scholar]
  • 45.Kaser A, Brandacher G, Steurer W, Kaser S, Offner FA, Zoller H, et al. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood. 2001;98(9):2720–2725. doi: 10.1182/blood.v98.9.2720. [DOI] [PubMed] [Google Scholar]
  • 46.Wang XH, Hong X, Zhu L, Wang YT, Bao JP, Liu L, et al. Tumor necrosis factor alpha promotes the proliferation of human nucleus pulposus cells via nuclear factor-κB, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase. Exp Biol Med (Maywood) 2015;240(4):411–417. doi: 10.1177/1535370214554533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zeng FC, Zeng MQ, Huang L, Li YL, Gao BM, Chen JJ, et al. Downregulation of VEGFA inhibits proliferation, promotes apoptosis, and suppresses migration and invasion of renal clear cell carcinoma. Onco Targets Ther. 2016;9:2131–2141. doi: 10.2147/OTT.S98002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lewis AM, Varghese S, Xu H, Alexander HR. Interleukin-1 and cancer progression: the emerging role of interleukin-1 receptor antagonist as a novel therapeutic agent in cancer treatment. J Transl Med. 2006;4:48. doi: 10.1186/1479-5876-4-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Paiva P, Salamonsen LA, Manuelpillai U, Walker C, Tapia A, Wallace EM, et al. Interleukin-11 promotes migration, but not proliferation, of human trophoblast cells, implying a role in placentation. Endocrinology. 2007;148:5566–5572. doi: 10.1210/en.2007-0517. [DOI] [PubMed] [Google Scholar]
  • 50.Shao J, Zhang B, Yu JJ, Wei CY, Zhou WJ, Chang KK, et al. Macrophages promote the growth and invasion of endometrial stromal cells by downregulating IL-24 in endometriosis. Reproduction. 2016;152(6):673–682. doi: 10.1530/REP-16-0278. [DOI] [PubMed] [Google Scholar]
  • 51.Brandacher G, Winkler C, Schroecksnadel K, Margreiter R, Fuchs D. Antitumoral activity of interferon-gamma involved in impaired immune function in cancer patients. Curr Drug Metab. 2006;7(6):599–612. doi: 10.2174/138920006778017768. [DOI] [PubMed] [Google Scholar]
  • 52.Veevers-Lowe J, Ball SG, Shuttleworth A, Kielty CM. Mesenchymal stem cell migration is regulated by fibronectin through alpha5beta1-integrin-mediated activation of PDGFR-β and potentiation of growth factor signals. J Cell Sci. 2011;124:1288–1300. doi: 10.1242/jcs.076935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Frijns CJ, Kappelle LJ. Inflammatory cell adhesion molecules in ischemic cerebrovascular disease. Stroke. 2002;33:2115–2122. doi: 10.1161/01.str.0000021902.33129.69. [DOI] [PubMed] [Google Scholar]
  • 54.Mostafa Mtairag E, Chollet-Martin S, Oudghiri M, Laquay N, Jacob MP, Michel JB, et al. Effects of interleukin-10 on monocyte/endothelial cell adhesion and MMP-9/TIMP-1 secretion. Cardiovasc Res. 2001;49:882–890. doi: 10.1016/s0008-6363(00)00287-x. [DOI] [PubMed] [Google Scholar]
  • 55.Shah AK, Lazatin J, Sinha RK, Lennox T, Hickok NJ, Tuan RS. Mechanism of BMP-2 stimulated adhesion of osteoblastic cells to titanium alloy. Biol Cell. 1999;91:131–142. doi: 10.1016/s0248-4900(99)80037-9. [DOI] [PubMed] [Google Scholar]
  • 56.Zhang J, Patel JM. Role of the CX3CL1-CX3CR57 axis in chronic inflammatory lung diseases. Int J Clin Exp Med. 2010;3:233–244. [PMC free article] [PubMed] [Google Scholar]
  • 57.Liu X, Xu X, Deng W, Huang M, Wu Y, Zhou Z, et al. CCL18 enhances migration, invasion and EMT by binding CCR58 in bladder cancer cells. Mol Med Rep. 2019;19:1678–1686. doi: 10.3892/mmr.2018.9791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Im KS, Graef AJ, Breen M, Lindblad-Toh K, Modiano JF, Kim JH. Interactions between CXCR59 and CXCL12 promote cell migration and invasion of canine hemangiosarcoma. Vet Comp Oncol. 2017;15:315–327. doi: 10.1111/vco.12165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Xu X, Huang P, Yang B, Wang X, Xia J. Roles of CXCL5 on migration and invasion of liver cancer cells. J Transl Med. 2014;12:193. doi: 10.1186/1479-5876-12-193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mori M, Morris SC, Orekhova T, Marinaro M, Giannini E, Finkelman FD. IL-4 promotes the migration of circulating B cells to the spleen and increases splenic B cell survival. J Immunol. 2000;164:5704–5712. doi: 10.4049/jimmunol.164.11.5704. [DOI] [PubMed] [Google Scholar]
  • 61.Ferland C, Flamand N, Davoine F, Chakir J, Laviolette M. IL-16 activates plasminogen-plasmin system and promotes human eosinophil migration into extracellular matrix via CCR62-chemokine-mediated signaling and by modulating CD4 eosinophil expression. J Immunol. 2004;173:4417–4424. doi: 10.4049/jimmunol.173.7.4417. [DOI] [PubMed] [Google Scholar]
  • 62.Panneerselvam J, Jin J, Shanker M, Lauderdale J, Bates J, Wang Q, et al. IL-24 inhibits lung cancer cell migration and invasion by disrupting the SDF-1/CXCR63 signaling axis. PLoS One. 2015;10(3):e0122439. doi: 10.1371/journal.pone.0122439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wolczyk D, Zaremba-Czogalla M, Hryniewicz-Jankowska A, Tabola R, Grabowski K, Sikorski AF, et al. TNF-α promotes breast cancer cell migration and enhances the concentration of membrane-associated proteases in lipid rafts. Cell Oncol (Dordr) 2016;39:353–363. doi: 10.1007/s13402-016-0280-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jin H, Pi J, Huang X, Huang F, Shao W, Li S, et al. BMP2 promotes migration and invasion of breast cancer cells via cytoskeletal reorganization and adhesion decrease: an AFM investigation. Appl Microbiol Biotechnol. 2012;93:1715–1723. doi: 10.1007/s00253-011-3865-3. [DOI] [PubMed] [Google Scholar]
  • 65.Hsu CJ, Wu MH, Chen CY, Tsai CH, Hsu HC, Tang CH, et al. AMP-activated protein kinase activation mediates CCL3-induced cell migration and matrix metalloproteinase-2 expression in human chondrosarcoma. Cell Commun Signal. 2013;11:68. doi: 10.1186/1478-811X-11-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lee S, Lee E, Ko E, Ham M, Lee HM, Kim ES, et al. Tumor-associated macrophages secrete CCL2 and induce the invasive phenotype of human breast epithelial cells through upregulation of ERO1-α and MMP-9. Cancer Lett. 2018;437:25–34. doi: 10.1016/j.canlet.2018.08.025. [DOI] [PubMed] [Google Scholar]
  • 67.Sun W, Liu DB, Li WW, Zhang LL, Long GX, Wang WF, et al. Interleukin-6 promotes the migration and invasion of nasopharyngeal carcinoma cell lines and upregulates the expression of MMP-2 and MMP-9. Int J Oncol. 2014;44:1551–1560. doi: 10.3892/ijo.2014.2323. [DOI] [PubMed] [Google Scholar]
  • 68.Bachelder RE, Wendt MA, Mercurio AM. Vascular endothelial growth factor promotes breast carcinoma invasion in an autocrine manner by regulating the chemokine receptor CXCR69. Cancer Res. 2002;62:7203–7206. [PubMed] [Google Scholar]
  • 69.Paiva P, Salamonsen LA, Manuelpillai U, Dimitriadis EP. Interleukin 11 inhibits human trophoblast invasion indicating a likely role in the decidual restraint of trophoblast invasion during placentation. Biol Reprod. 2009;80:302–310. doi: 10.1095/biolreprod.108.071415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.de Jager SC, Bot I, Kraaijeveld AO, Korporaal SJ, Bot M, van Santbrink PJ, et al. Leukocyte-specific CCL3 deficiency inhibits atherosclerotic lesion development by affecting neutrophil accumulation. Arterioscler Thromb Vasc Biol. 2013;33:e75–e83. doi: 10.1161/ATVBAHA.112.300857. [DOI] [PubMed] [Google Scholar]
  • 71.Mehraj V, Ramendra R, Isnard S, Dupuy FP, Lebouché B, Costiniuk C, et al. CXCL13 as a biomarker of immune activation during early and chronic HIV infection. Front Immunol. 2019;10:289. doi: 10.3389/fimmu.2019.00289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Marshall A, Celentano A, Cirillo N, McCullough M, Porter S. Tissue-specific regulation of CXCL9/10/11 chemokines in keratinocytes: Implications for oral inflammatory disease. PLoS One. 2017;12:e0172821. doi: 10.1371/journal.pone.0172821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ma Xiaojing, Yan Wenjun, Zheng Hua, Du Qinglin, Zhang Lixing, Ban Yi, Li Na, Wei Fang. Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells. F1000Research. 2015;4:1465. doi: 10.12688/f1000research.7010.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Park S, Cheon S, Cho D. The dual effects of interleukin-18 in tumor progression. Cell Mol Immunol. 2007;4:329–335. [PubMed] [Google Scholar]
  • 75.Shen J, James AW, Zara JN, Asatrian G, Khadarian K, Zhang JB, et al. BMP2-induced inflammation can be suppressed by the osteoinductive growth factor NELL-1. Tissue Eng Part A. 2013;19(21–22):2390–2401. doi: 10.1089/ten.tea.2012.0519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hermann JA, Hall MA, Maini RN, Feldmann M, Brennan FM, et al. Important immunoregulatory role of interleukin-11 in the inflammatory process in rheumatoid arthritis. Arthritis Rheum. 1998;41:1388–1397. doi: 10.1002/1529-0131(199808)41:8<1388::AID-ART7>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 77.Wood S, Jayaraman V, Huelsmann EJ, Bonish B, Burgad D, Sivaramakrishnan G, et al. Pro-inflammatory chemokine CCL2 (MCP-1) promotes healing in diabetic wounds by restoring the macrophage response. PLoS One. 2014;9:e91574. doi: 10.1371/journal.pone.0091574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Damås JK, Landrø L, Fevang B, Heggelund L, Tjønnfjord GE, Fløisand Y, et al. Homeostatic chemokines CCL19 and CCL21 promote inflammation in human immunodeficiency virus-infected patients with ongoing viral replication. Clin Exp Immunol. 2009;157(3):400–407. doi: 10.1111/j.1365-2249.2009.03976.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.De Plaen IG, Han XB, Liu X, Hsueh W, Ghosh S, May MJ. Lipopolysaccharide induces CXCL2/macrophage inflammatory protein-2 gene expression in enterocytes via NF-kappaB activation: independence from endogenous TNF-alpha and platelet-activating factor. Immunology. 2006;118(2):153–163. doi: 10.1111/j.1365-2567.2006.02344.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hartgring SA, Willis CR, Bijlsma JW, Lafeber FP, van Roon JA. Interleukin-7-aggravated joint inflammation and tissue destruction in collagen-induced arthritis is associated with T-cell and B-cell activation. Arthritis Res Ther. 2012;14:R137. doi: 10.1186/ar3870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mathy NL, Scheuer W, Lanzendörfer M, Honold K, Ambrosius D, Norley S, et al. Interleukin-16 stimulates the expression and production of pro-inflammatory cytokines by human monocytes. Immunology. 2000;100:63–69. doi: 10.1046/j.1365-2567.2000.00997.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Etemadi N, Holien JK, Chau D, Dewson G, Murphy JM, Alexander WS, et al. Lymphotoxin alpha induces apoptosis, necroptosis and inflammatory signals with the same potency as tumour necrosis factor. FEBS J. 2013;280:5283–5297. doi: 10.1111/febs.12419. [DOI] [PubMed] [Google Scholar]
  • 83.Ham B, Fernandez MC, D’Costa Z, Brodt P. The diverse roles of the TNF axis in cancer progression and metastasis. Trends Cancer Res. 2016;11:1–27. [PMC free article] [PubMed] [Google Scholar]
  • 84.Linker RA, Mäurer M, Gaupp S, Martini R, Holtmann B, Giess R, et al. CNTF is a major protective factor in demyelinating CNS disease: a neurotrophic cytokine as modulator in neuroinflammation. Nat Med. 2002;8(6):620–624. doi: 10.1038/nm0602-620. [DOI] [PubMed] [Google Scholar]
  • 85.Alikhan MA, Jones CV, Williams TM, Beckhouse AG, Fletcher AL, Kett MM, et al. Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. Am J Pathol. 2011;179:1243–1256. doi: 10.1016/j.ajpath.2011.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Viel S, Marçais A, Guimaraes FS, Loftus R, Rabilloud J, Grau M, et al. TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal. 2016;9:ra19. doi: 10.1126/scisignal.aad1884. [DOI] [PubMed] [Google Scholar]
  • 87.Allen F, Rauhe P, Askew D, Tong AA, Nthale J, Eid S, et al. CCL3 enhances antitumor immune priming in the lymph node via IFNgamma with dependency on natural killer cells. Front Immunol. 2017;8:1390. [DOI] [PMC free article] [PubMed]
  • 88.Ma Hailong, Jin Shufang, Yang Wenyi, Tian Zhuowei, Liu Shuli, Wang Yang, Zhou Ge, Zhao Mei, Gvetadze Shalva, Zhang Zhiyuan, Hu Jingzhou. Interferon-α Promotes the Expression of Cancer Stem Cell Markers in Oral Squamous Cell Carcinoma. Journal of Cancer. 2017;8(12):2384–2393. doi: 10.7150/jca.19486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lee HL, Park MH, Song JK, Jung YY, Kim Y, Kim KB, et al. Tumor growth suppressive effect of IL-4 through p21-mediated activation of STAT6 in IL-4Rα overexpressed melanoma models. Oncotarget. 2016;7:23425–23438. doi: 10.18632/oncotarget.8111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Qin Z, van Tits LJ, Buurman WA, Blankenstein T. Human lymphotoxin has at least equal antitumor activity in comparison to human tumor necrosis factor but is less toxic in mice. Blood. 1995;85(10):2779–2785. [PubMed] [Google Scholar]
  • 91.Hofmann A, Blau HM. Death of solid tumor cells induced by Fas ligand expressing primary myoblasts. Somat Cell Mol Genet. 1997;23:249–257. doi: 10.1007/BF02674416. [DOI] [PubMed] [Google Scholar]
  • 92.Razidlo GL, Burton KM, McNiven MA. Interleukin-6 promotes pancreatic cancer cell migration by rapidly activating the small GTPase CDC42. J Biol Chem. 2018;293:11143–11153. doi: 10.1074/jbc.RA118.003276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Silva A, Laranjeira AB, Martins LR, Cardoso BA, Demengeot J, Yunes JA, et al. IL-7 contributes to the progression of human T-cell acute lymphoblastic leukemias. Cancer Res. 2011;71:4780–4789. doi: 10.1158/0008-5472.CAN-10-3606. [DOI] [PubMed] [Google Scholar]
  • 94.Ara T, Declerck YA. Interleukin-6 in bone metastasis and cancer progression. Eur J Cancer. 2010;46:1223–1231. doi: 10.1016/j.ejca.2010.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhu Z, Zhang X, Guo H, Fu L, Pan G, Sun Y. CXCL13-CXCR96 axis promotes the growth and invasion of colon cancer cells via PI3 K/AKT pathway. Mol Cell Biochem. 2015;400(1–2):287–295. doi: 10.1007/s11010-014-2285-y. [DOI] [PubMed] [Google Scholar]
  • 96.Li H, Meng YH, Shang WQ, Liu LB, Chen X, Yuan MM, et al. Chemokine CCL24 promotes the growth and invasiveness of trophoblasts through ERK1/2 and PI3K signaling pathways in human early pregnancy. Reproduction. 2015;150:417–427. doi: 10.1530/REP-15-0119. [DOI] [PubMed] [Google Scholar]
  • 97.Hu W, Liu Y, Zhou W, Si L, Ren L. CXCL16 and CXCR98 are coexpressed in human lung cancer in vivo and mediate the invasion of lung cancer cell lines in vitro. PLoS One. 2014;9:e99056. doi: 10.1371/journal.pone.0099056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.DeWitt J, Pappas A, Nishi R. Ciliary neurotrophic factor reduces the proliferation and promotes the differentiation of TH-MYCN transformed sympathoadrenal progenitors. Dev Neurosci. 2014;36:422–431. doi: 10.1159/000365281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang MH, Zhou XM, Zhang MY, Shi L, Xiao RW, Zeng LS, et al. BMP2 promotes proliferation and invasion of nasopharyngeal carcinoma cells via mTORC1 pathway. Aging (Albany NY) 2017;9(4):1326–1340. doi: 10.18632/aging.101230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zong M, Lu T, Fan S, Zhang H, Gong R, Sun L, et al. Glucose-6-phosphate isomerase promotes the proliferation and inhibits the apoptosis in fibroblast-like synoviocytes in rheumatoid arthritis. Arthritis Res Ther. 2015;17:100. doi: 10.1186/s13075-015-0619-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Verheul HM, Jorna AS, Hoekman K, Broxterman HJ, Gebbink MF, Pinedo HM, et al. Vascular endothelial growth factor-stimulated endothelial cells promote adhesion and activation of platelets. Blood. 2000;96(13):4216–4221. [PubMed] [Google Scholar]

Articles from Tissue Engineering and Regenerative Medicine are provided here courtesy of Springer

RESOURCES