Open Access

Modulation of the secretion of mesenchymal stem cell immunoregulatory factors by hydrolyzed fish collagen

  • Authors:
    • Chao Liu
    • Jiao Sun
  • View Affiliations

  • Published online on: April 22, 2020     https://doi.org/10.3892/etm.2020.8674
  • Pages: 375-384
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The aim of the present study was to investigate the possible immunomodulatory effects of osteogenically differentiated bone marrow mesenchymal stem cells induced by hydrolyzed fish collagen. Marine biomaterials have attracted significant attention for their environmental friendliness and renewability. Hydrolyzed fish collagen (HFC) has been discovered to induce the osteoblastic differentiation of stem cells, which underlies the foundation for its application in tissue engineering. Stem cells and their biomaterial carriers face acute immune rejection mediated by host macrophages. A potential strategy for combatting rejection in stem cell therapy is to modify the polarization of macrophages. However, whether HFC‑induced mesenchymal stem cells maintain their immunomodulatory ability remains to be determined. To understand this phenomenon, a co‑culture model of direct contact was established between bone marrow mesenchymal stem cells (BMSCs) and RAW264.7 macrophages, where the secretion of nitrous oxide from macrophages was measured using Griess colorimetric assay. ELISAs were performed to measure the secretion of interleukin (IL)‑1β, IL‑6, transforming growth factor (TGF)‑β and IL‑10, whilst reverse transcription‑quantitative PCR was used to assess the expression levels of IL‑1β, IL‑6, CD206, resistin‑like molecule α (FIZZ1) and prostaglandin E2 receptor 4 (EP4). In addition, the expression levels of relevant proteins in the phosphorylated‑cyclic AMP‑responsive element‑binding protein‑CCAAT/enhancer‑binding protein β (EBPβ) pathway were investigated using western blotting. HFC‑induced BMSCs were found to suppress the expression levels of IL‑1β and IL‑6, whilst increasing the expression levels of CD206 and FIZZ1 in RAW264.7 macrophages. HFC‑induced BMSCs also inhibited the secretion of IL‑1β and IL‑6, whilst promoting the secretion of TGF‑β and IL‑10 secretion from RAW264.7 macrophages. Mechanistic studies using western blotting discovered that HFC stimulated the secretion of prostaglandin E2 from BMSCs, which subsequently increased the expression of EP4 on the macrophages. EP4 then increased the expression levels of C/EBPβ and arginase 1 further. In conclusion, results from the present study suggested that following induction with HFC, BMSCs maintain their immunomodulatory activity.

Introduction

With the technological advancement of stem cell and tissue engineering, regenerative medicine has become a hot topic in the field of biological medicine (1). The self-renewal and multidirectional differentiation of stem cells enables them to serve as seed cells for tissue engineering, facilitating the repair of damaged tissues or organs (2). In tissue engineering, ≥2 conditions are required to optimize the application of stem cells, including the presence of an effective inducer and tolerance to immunological assault (3). A number of biomaterials have been previously found to induce the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) (4), such as hydrolyzed fish collagen (HFC) (5). Since it is readily available and accessible, HFC is a material that warrants further investigations in this field.

The biological activity of HFC has become a notable focus of research. Blanco et al (6) revealed that HFC possessed significant antioxidant properties, whilst Liu and Sun (7) observed that HFC induced the osteogenic differentiation of human periodontal ligament cells in another study. HFC was found to induce adipose-derived stromal cell chondrogenesis as effectively as transforming growth factor (TGF)-β1(8), whereas the anti-inflammatory properties of HFC has also been previously documented (9). Collectively, these findings suggested that HFC possesses a number of biological activities with the potential for future clinical application.

Immunological rejection remains to be the primary limitation for the transplantation of allogenic stem cells and their derivatives (10). Stem cells and their associated scaffolds are known to face acute immune rejection mediated by host macrophages, hindering the migration of reparative cells and weakening the ability of new tissues to propagate to their surroundings, in turn leading to failure in tissue regeneration (11). Although previous research has largely focused on inhibiting the activation of host immunity (12,13), macrophages have also attracted increased attention for their substantial plasticity and phenotype-switching capacity (14). Owing to their prominent phenotypic plasticity, macrophages have been discovered to mediate both proinflammatory rejection and anti-inflammatory tissue remodeling (15). The exposure of M1 macrophages to M2 signals and vice versa, has been discovered to induce the re-polarization of differentiated macrophages, which demonstrates their high functional plasticity and potential therapeutic use (16). Therefore, modulating macrophage plasticity may provide a novel strategy for combating immune rejection in tissue engineering.

Mesenchymal stem cells (MSCs) possess unique immunoregulatory properties. A previous study reported that MSC transplantation modulated the immune response against allografts and alleviated transplant rejection, prolonging allograft survival (17). In addition, macrophages co-cultured with MSCs were found to consistently express high levels of CD206, a marker of alternatively-activated macrophages (18). In addition, the secretion levels of interleukin (IL)-10 and IL-12 were found to be increased and reduced, respectively, which is characteristic of alternatively-activated macrophages (19). In another previous study, macrophages co-cultured with MSCs were revealed to express lower levels of tumor necrosis factor α (TNF-α) compared with macrophages cultured alone, suggesting that MSCs modulate the inflammatory response by inducing M2 macrophage differentiation (20,21). Although HFC has been revealed to induce the osteogenic differentiation of MSCs as aforementioned, the effects of HFC on the immunomodulatory functions of MSCs remain unknown.

In the present study, a cell-cell contact co-culture model between BMSCs and macrophages was established to determine the regulatory effects of HFC-induced BMSCs on the crucial inflammatory factors associated with macrophages. Additionally, the immunomodulatory mechanism of BMSCs was investigated, providing a foundation for the application of HFC and BMSCs in tissue engineering.

Materials and methods

Materials

HFC was supplied by the Shanghai Fisheries Research Institute (Shanghai, China).

RAW264.7 cell culture

The murine macrophage RAW264.7 cell line was obtained from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. The macrophages were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 50 µg/ml streptomycin and 100 IU/ml penicillin, which were maintained at 37˚C (5% CO2) in a humidified atmosphere.

Isolation and in vitro culture of BMSCs

The present study was approved by the Ethics Committee of Shanghai Ninth People's Hospital, affiliated with the School of Medicine, Shanghai Jiao Tong University (Shanghai, China). The BMSCs originated from bone marrow mononuclear cells, which have the potential to differentiate into a number of different cell types, including osteoblasts, adipocytes, chondrocytes and neural cells (22). BMSCs have garnered considerable research attention due to their simplicity of preparation, ethical considerations, accessibility and low immunogenicity (23). In the present study, 10 male Sprague Dawley rats (age at sacrifice, 4 weeks; weight, 62.3±2.5 g) were sacrificed by cervical dislocation and the body was soaked in 75% ethanol for 5 min at room temperature. All rats were housed in a temperature-controlled room (21±2˚C) with relative air humidity of 40-60%, under a 12-h light/dark cycle, with free access to food and water. The tibia and femur of the rats were then obtained under sterile conditions at room temperature, where a 5 ml syringe and a 25-gauge needle was used to flush the bone marrow from the femur and tibia of rats by injecting 0.5 ml DMEM supplemented with 10% FBS, 100 IU/ml penicillin and 50 µg/ml streptomycin, into the bone marrow cavity three times. The washing fluid was collected and centrifuged at 200 x g at room temperature for 10 min, following which the supernatant was discarded. Subsequently, the pelleted cells were dispersed and centrifuged at 200 x g at room temperature for 10 min again and the supernatant was discarded.

BMSCs were subsequently cultured in low glucose DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) at 37˚C in 5% CO2. thereafter, with the medium being changed every day for the first 3 days. At 80% confluence, the cells were passaged into fresh plates by trypsinization, where third generation BMSCs were collected for follow-up experiments. The morphology of the primary BMSCs was observed under a phase contrast microscope (Magnification, x10; Nikon Corporation).

For osteogenic and adipogenic differentiation, 1x105 BMSCs/ml were seeded into six-well plates in the low glucose DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.). After reaching 80% confluence, the cells were treated with osteogenic differentiation medium (DMEM supplemented with 10% FBS, 50 ng/ml ascorbic acid, 100 µmol/l dexamethasone and 10 mmol/l β-glycerophosphate (All from Sigma-Aldrich; Merck KGaA) or adipogenic differentiation medium [DMEM supplemented with 10% FBS, 0.5 mM 3-isobutyl-1-methylxanthine (Sigma-Aldrich; Merck KGaA), 50 µM indomethacin (Sigma-Aldrich; Merck KGaA), 10 µM dexamethasone Sigma-Aldrich; Merck KGaA) and 10 µg/ml insulin Sigma-Aldrich; Merck KGaA)] for 14 days at 37 ˚C, where the culture medium was changed every 3 days.

Establishment of the cell co-culture system

BMSCs were treated with either 0.2 mg/ml HFC for 7 days at 37˚C or with 0.2 mg/ml HFC for 7 days followed by 10 µM NS-398 (Tocris Bioscience), a specific cyclooxygenase 2 (COX-2) inhibitor, for 1 day at 37˚C. The cells were then plated into 24-well plates at a density of 5x104/ml. Following 24 h of incubation at 37˚C, RAW264.7 macrophages were pre-stimulated with lipopolysaccharide (1 µg/ml; Sigma-Aldrich; Merck KGaA) for 30 min at 37˚C and then added to the plates containing the BMSCs at a density of 1x104 cells/ml. RAW264.7 macrophages cultured alone were used as a control. The cells and supernatants were collected following incubation for 24 h at 37˚C in 5% CO2.

MTT assay

At 24 h after co-culture initiation, cell viability was determined for all experimental groups using an MTT assay. Briefly, 400 µl MTT solution was added to each well and incubated for 4 h at 37˚C. Following the removal of medium, 200 µl DMSO was added to each well and incubated for a further 10 min at room temperature. The optical density value of each well was measured using a plate reader at 570 nm.

Determination of nitrous oxide (NO) concentration in cell supernatants

The supernatants from each group were collected, where the concentration of NO in the supernatant was determined using Griess' method (cat. no. S0021; Beyotime Institute of Biotechnology), according to the manufacturer's protocol.

ELISAs

The concentrations of IL-1β (cat. no. SMLB00C), IL-6 (cat. no. SM6000B), TGF-β (cat. no. DY1679) and IL-10 (cat. no. SM1000B) in the co-culture medium were measured in the supernatants using corresponding ELISA kits (R&D Systems, Inc.) according to the manufacturer's protocols. To analyze the concentration of prostaglandin E2 (PGE2), the supernatants of the treated BMSCs were collected before the RAW264.7 macrophages were added, where the concentration was determined using an ELISA kit (cat. no. MBS262150; MyBioSource, Inc.), according to the manufacturer's protocol.

Reverse transcription-quantitative PCR (RT-qPCR)

The cells were separated using magnetic beads as previously described (24). CD34 (10 µg; cat. no. ab187282; Abcam), CD45 (10 µg; cat. no. ab25078; Abcam) and Dynabeads™ Goat Anti-Mouse IgG beads (cat. no. 11033; Invitrogen; Thermo Fisher Scientific, Inc.) were used according to the manufacturer's protocol. In RAW264.7 macrophages, the expression levels of IL-1β, IL-6, CD206, resistin-like α (FIZZ1) and prostaglandin E2 receptor 4 (EP4) were analyzed. In BMSCs, IL-1β, IL-6, CD206, FIZZ1, runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), alkaline phosphatase (ALP), lipoprotein lipase (LPL), adipose differentiation related protein (ADRP) and peroxisome proliferator-activated receptor γ (PPARγ) expression levels were assessed. Total RNA was extracted from RAW264.7 macrophages or BMSCs using RNAeasy™ Animal RNA Isolation Kit (cat. no. R0024FT; Beyotime Institute of Biotechnology) according to the manufacturer's protocol, the total RNA (10 µg) was then reverse transcribed into cDNA using the PrimeScript™ RT Reagent kit (Takara Bio, Inc.), according to the manufacturer's protocol, the reverse transcription reaction condition was as follows: 37˚C for 15 min, 85˚C for 5 sec and 4˚C for 2 min. qPCR was subsequently performed using the Power SYBR™ Green Master Mix according to manufacturer's protocol (cat. no. 4368577; Thermo Fisher Scientific, Inc.), The thermal cycling conditions consisted of an initial denaturation at 95˚C for 3 min, followed by 40 cycles of denaturation at 95˚C, 15 sec, annealing 60˚C for 30 sec and elongation 72˚C for 30 sec. The primer sequences used for qPCR are listed in Table I. Target gene expression was quantified using the 2-ΔΔCq method and normalized to that of the GAPDH gene (25).

Table I

Sequences of primers used for reverse transcription-quantitative PCR.

Table I

Sequences of primers used for reverse transcription-quantitative PCR.

GenePrimer sequence (5'→3')
Runt-related transcription factor 2F: GCCGGGAATGATGAGAACTA
 R: GGACCGTCCACTGTCACTTT
OsteocalcinF: TGCATTCTGCCTCTCTGACC
 R: ACCACCTTACTGCCCTCCTG
Alkaline phosphataseF: AAGGCTTCTTCTTGCTGGTG
 R: GCCTTACCCTCATGATGTCC
Peroxisome proliferator-activated receptor γF: CCAAGTGACTCTGCTCAAGTATGG
 R: CATGAATCCTTGTCCCTCTGATATG
Lipoprotein lipaseF: TGAAGACACAGCTGAGGACA
 R: GATCACCACAAAGGTTTTGC
Adipose differentiation related proteinF: ATTCTGGACCGTGCCGATT
 R: CTGCTACTGATGCCATTTTTCCT
IL-1βF: GGACAGAATATCAACCAACAAGTGATA
 R: GTGTGCCCGTCTTTCATTACACAG
IL-6F: CCAGAAACCGCTATGAAGTTCCT
 R: CACCAGCATCAGTCCCAAGA
CD206F: GTCTGAGTGTACGCAGTGGTTGG
 R: TCTGATGATGGACTTCCTGGTAGCC
Resistin-like αF: TGCTGGGATGACTGCTACTG
 R: TGCTGGGATGACTGCTACTG
Prostaglandin E2 receptor 4F: TCTACTTGCTCCCAGTGGACATAGATGG
 R: GAACAGACTCCTGAACTGGGTATGGTTC
GAPDHF: AGGTGAAGGTCGGAGTCAACG
 R: CCTGGAAGATGGTGATGGGAT

[i] IL, interleukin; F, forward; R, reverse.

Western blotting

The cells were separated using the magnetic beads method as aforementioned and previously described (24). Total protein was extracted from RAW264.7 macrophages or BMSCs using RIPA lysis buffer (Beyotime Institute of Biotechnology) with a protease inhibitor cocktail (Roche Diagnostics). Total protein was quantified using a bicinchoninic acid assay kit (Pierce; Thermo Fisher Scientific, Inc.) and 20 µg protein/lane was separated via 12% SDS-PAGE. The separated proteins were subsequently transferred onto a polyvinylidene fluoride membrane and blocked with 5% skim milk for 2 h at room temperature. The membranes were further incubated overnight at 4˚C with the following mouse primary antibodies for the RAW264.7 protein samples: Anti-cyclic AMP-responsive element-binding protein (CREB) (1:1,000; cat. no. ab31387; Abcam), anti-phosphorylated (p)-CREB (1:500; cat. no. ab32096; Abcam), anti-CCAAT/enhancer-binding protein (C/EBP) (1:1,000; cat. no. ab40764; Abcam), anti-arginase 1 (Arg-1; 1:500; cat. no. 93668; Cell Signaling Technology, Inc.), anti-inducible nitric oxide synthase (iNOS; 1:1,000; cat. no. ab178945; Abcam), and anti-β-actin (1:1,000; cat. no. ab8227; Abcam). The membranes were incubated with the following rat primary antibodies for the BMSC protein samples at room temperature for 1 h: Anti-CD29 (1:500; cat. no. AF2405, R&D Systems, Inc.), anti-CD90 (1:1,000; cat. no. ab92574; Abcam), anti-CD34 (1:1,000 dilution; cat. no. AF4117; R&D Systems, Inc.), anti-CD45 (1:1,000; cat. no. ab10558; Abcam) and anti-β-actin (1:1,000; cat. no. ab8227; Abcam). Following the primary antibody incubation, the membranes were washed with 0.1% TBS-Tween 3 times for 10 min each prior to incubation with a horseradish peroxidase-conjugated secondary antibodies at room temperature for 1 h [(cat. no. ab6802; 1:5,000; Abcam) or (cat. no. ab6885; 1:5,000; Abcam)]. The protein bands were visualized using ECL reagents (EMD Millipore) and quantified using ImageJ software (version 1.48; National Institutes of Health).

Statistical analysis

All experiments were repeated three times and all data in this study were presented as the mean ± SD. Statistical analysis was performed using SPSS 22.0 software (IBM Corp.). Statistical comparisons were made using one-way ANOVA and a Tukey's post hoc test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Characterization of BMSCs

The isolated cells were confirmed to be BMSCs based on their spindle-shaped morphology and adherence properties (Fig. 1A). In addition, western blotting confirmed the expression of CD29 and CD90 and the lack of CD34 and CD45 expression of BMSCs (Fig. 1B), consistent with the previous studies that BMSCs express CD29 and CD90, but not CD34 and CD45 (26,27). The expression levels of osteogenic (RUNX2, ALP and OCN) and adipogenic (LPL, ADRP and PPARγ) markers were also significantly increased in osteogenic- or adipogenic-induced BMSCs in vitro compared with those in unstimulated BMSCs (Fig. 1C and D).

Cell viability

Viability in the co-culture system was found to be comparable compared with that observed for RAW264.7 macrophages or BMSCs when either were cultured alone, since no significant differences were observed in cell viability between the co-culture and the monocultures (Fig. 2), suggesting that co-culturing or HFC does not negatively influence the viability of RAW264.7 macrophages and BMSCs.

NO production

Co-culturing with HFC-induced BMSCs was revealed to significantly reduce NO production by RAW264.7 macrophages compared with that by RAW264.7 macrophages when cultured alone (Fig. 3). NO production in the co-culture system consisting of untreated BMSCs and RAW264.7 macrophages was also found to be significantly decreased compared with that by RAW264.7 macrophages alone (Fig. 3). However, no significant difference was observed between the RAW264.7 co-cultured with unstimulated BMSCs and RAW264.7 co-cultured with HFC-induced BMSCs (Fig. 3).

Cytokine secretion

The concentrations of IL-1β, IL-6, TGF-β and IL-10 in the RAW264.7 macrophage supernatants were measured using ELISAs. The secretion of IL-1β and IL-6 from the RAW264.7 macrophages co-cultured with HFC-induced BMSCs was found to be significantly decreased compared with that from the RAW264.7 group when cultured alone (Fig. 4A and B). By contrast, the concentrations of TGF-β and IL-10 in the supernatants of RAW264.7 macrophages co-cultured with HFC-induced BMSCs were significantly increased compared with those found in those of the RAW264.7 monoculture (Fig. 4C and D). Notably, the concentration of IL-10 in the supernatants of the HFC-induced BMSC + RAW264.7 group was also significantly increased compared with that in the untreated BMSC + RAW264.7 group (Fig. 4D). However, a significant difference in the concentrations of IL-1β, IL-6 or TGF-β between the untreated or HFC-induced BMSC + RAW264.7 groups was not observed.

Expression of genes associated with inflammation in RAW264.7 macrophages and BMSCs

IL-1β and IL-6 are important M1-type macrophage markers (28). RT-qPCR results demonstrated that HFC-induced BMSCs significantly reduced the expression levels of IL-1β and IL-6 mRNA in RAW264.7 macrophages compared with those in the RAW264.7 macrophage monoculture. In addition, there were no significant differences in RAW264.7 macrophages co-cultured with either HFC-induced BMSCs or untreated BMSCs (Fig. 5A and B). Conversely, HFC-induced BMSCs increased the expression levels of CD206 and FIZZ1 mRNA in RAW264.7 macrophages compared with those in the RAW264.7 macrophages alone (Fig. 5C and D), which are important M2-type macrophage markers (29). Notably, the mRNA expression levels of IL-1β and IL-6 in BMSCs alone were found to be significantly decreased compared with those in RAW264.7 macrophages alone (Fig. 5A and B). No CD206 and FIZZ1 mRNA expression could be detected in BMSCs (Fig. 5C and D).

Measurement of PGE2 secretion by BMSCs and EP4 expression in RAW264.7 macrophages

ELISA results revealed that HFC treatment significantly increased the secretion of PGE2 in BMSCs compared with that by untreated BMSCs, which was antagonized by NS-398, a specific inhibitor of COX-2, which is responsible for PGE2 production (Fig. 6A) (30). EP4 is the receptor for PGE2, an important transmembrane G protein-coupled receptor in macrophages (31). The results of the RT-qPCR analysis demonstrated that HFC-induced BMSCs significantly increased the expression levels of EP4 mRNA compared with RAW264.7 alone. The stimulatory effect of increased EP4 expression following co-culture with HFC-induced BMSCs was significantly greater compared with those in the co-culture consisting of macrophages and untreated BMSCs (Fig. 6B). Notably, the effects induced by HFC-induced BMSCs were significantly reversed in the presence of NS-398 (Fig. 6B).

Effect of HFC-induced BMSCs on the expression levels of proteins associated with the CREB pathway in RAW264.7 macrophages

Western blotting data suggested that HFC-induced BMSCs significantly increased CREB phosphorylation in the macrophages compared with that in all other groups, especially when compared with that in the BMSCs + RAW264.7 group (Fig. 7A and B), resulting in the increased expression levels of the C/EBPβ protein (Fig. 7A and C), upregulation of the Arg-1 protein and the inhibition of iNOS expression compared with those in all other groups, especially when compared with those in the BMSCs + RAW264.7 group (Fig. 7A and D). These aforementioned effects were all found to be significantly reversed following the application of NS-398 (Fig. 7).

Discussion

In close proximity, cells communicate via paracrine signaling or cell-cell contact. Direct-contact co-culture systems cover both of these aspects; therefore, they can be considered to be more representative of the cellular microenvironment in vivo compared with monoculture systems, which can be used to study cell-cell interactions effectively in vitro (32). To date, studies on the immunomodulatory effects of BMSCs have primarily focused on interactions with T and B lymphocytes, natural killer and dendritic cells (33-36), but those on macrophages remain insufficient.

Since NO is considered the most sensitive and efficient indicator of inflammatory macrophages (37), it was analyzed in the present study. HFC-induced BMSCs were found to significantly inhibit the production of NO in the co-culture system, with this effect was comparable with the untreated BMSCs, suggesting that the HFC-induced BMSCs had retained their immunomodulatory functions.

Macrophages serve an important role in immunomodulation by secreting inflammatory factors, where the polarization state of the macrophages can be identified by changes to the cytokine profile (38). For example, M1 macrophages primarily secrete proinflammatory cytokines IL-1β, IL-6 and TNF-α (39), whilst M2 macrophages secrete vascular endothelial growth factor, TGF-β, endothelial growth factor and IL-10, all of which are involved in anti-inflammatory responses and tissue regeneration (16). To confirm the effect of HFC-induced BMSCs on macrophage polarization in the present study, the secretion of the relevant inflammatory factors was also analyzed. The results revealed that HFC-induced BMSCs inhibited the secretion of IL-1β and IL-6, whilst promoting the secretion of TGF-β and IL-10 by RAW246.7 macrophages. IL-1β and IL-6 are well-recognized proinflammatory cytokines and important markers of M1 macrophages. IL-1β is derived from macrophages and serve as the primary regulator of innate immune and inflammatory responses (40). By contrast, IL-6, which is predominantly secreted by T cells and macrophages, is an important member of the inflammatory network (41). As a multifunctional cytokine, IL-6 has been discovered to regulate cellular immune responses, inflammation and hematopoiesis (41). Since macrophages are one of the main sources of TGF-β, they serve a broader role in cellular proliferation, differentiation and immune functioning (42). TGF-β inhibits the proliferation of immune effector cells and the generation of cytotoxic lymphocytes from CD8+ cells, in addition to directly inhibiting T-helper cell differentiation (43). IL-10 is an anti-inflammatory cytokine that can directly inhibit the activation of inflammatory cells, thereby reducing the production of inflammatory cytokines (44). The timely and moderate production of IL-10 has also been discovered to relieve inflammation and protect normal tissues from inflammatory injuries (45). In the present study, HFC-induced BMSCs were found to inhibit the secretion of IL-1β and IL-6, whilst increasing the levels of TGF-β and IL-10 secretion, suggesting that HFC treatment did not impair the immunomodulatory functions of BMSCs.

The immunomodulatory role of HFC-induced BMSCs was subsequently investigated on genetic level. HFC-induced BMSCs inhibited the expression levels of IL-1β and IL-6 mRNA, whilst increasing the expression levels of CD206 and FIZZ1 in the macrophages. Both CD206 and FIZZ1 are M2 macrophage markers (46), therefore, these results indicated that HFC-induced BMSCs may regulate macrophage polarization on transcriptional level.

A previous study demonstrated that PGE2 also regulated macrophage polarization (47). To verify whether HFC-induced BMSCs exerted their activity via PGE2, its secretion by BMSCs was investigated in the present study. HFC treatment was found to induce the production of PGE2 in BMSCs, which was reversed by the specific PGE2 inhibitor NS-398. It has been suggested that the regulatory role of PGE2 is mediated via the EP4 receptor on macrophages (48). Consistent with these findings, results of the current study revealed that the expression levels of EP4 were increased in macrophages following co-culture with HFC-induced BMSCs, which were reversed by the presence of NS-398. These results suggested that the immunomodulatory role of HFC-induced BMSCs may be mediated through PGE2.

After identifying the important immunoregulatory role of PGE2 in BMSCs, the question of how PGE2 inhibited M1-polarization whilst promoting M2 polarization was raised. To clarify the underlying mechanism of PGE2, potential pathways were studied using western blotting. The results discovered that the phosphorylation levels of CREB and the protein expression levels of C/EBPβ were significantly increased in the presence of HFC-induced BMSCs, which ultimately increased the expression of Arg-1 and inhibited the production of iNOS. By contrast, in the presence of NS-398, the expression levels of C/EBPβ, Arg-1 and CREB phosphorylation were inhibited, whereas the expression levels of iNOS were increased. These findings indicated that HFC may induce the production of PGE2 by BMSCs, which may subsequently activate the transcription of Arg-1 in macrophages by activating the p-CREB/C/EBPβ pathway, resulting in polarization towards the M2 subtype.

It is important to differentiate the two different cell types in the co-culture system, as both BMSCs and RAW264.7 macrophages produce NO and express a number of inflammation-associated genes, including IL-1β and IL-6. In the present study, a sufficient and suitable control group was used to achieve this. The BMSC + RAW264.7 co-culture group was used as a control for NO production, whereas the BMSC + RAW264.7 co-culture and BMSC monoculture were used for mRNA expression, which confirmed that HFC-induced BMSCs were influencing these effects.

It should be noted that BMSCs are the most widely used cells in bone tissue engineering applications. The focus of the present study was on bone tissue engineering, which was the reason for BMSCs being used. In fact, co-culture of RAW264.7 macrophages with other types of cells, including hematopoietic stem cells or embryonic stem cells, would serve as an excellent control for comparative purposes to the effects of BMSCs, which will be investigated in future studies.

In conclusion, the findings of the present study suggested that HFC-induced MSCs may significantly inhibit the expression of M1-macrophage markers and promote the expression of M2 markers, indicating that HFC-induced BMSCs may exert significant immunomodulatory activities. Mechanistically, the findings indicated that HFC may promote the secretion of PGE2 from BMSCs, which may activate the p-CREB/C/EBPβ pathway through binding to the EP4 receptor on the macrophages, resulting in an increase in Arg-1 expression and a reduction in iNOS expression. The biological functions of HFC were clarified, which may facilitate further development of HFC-based biomaterials.

Acknowledgements

The authors would like to thank Professor Nanping Wang (Shanghai Fisheries Research Institute, Shanghai, China) for generously providing the HFC.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 31600760).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

CL performed all the experiments. JS interpreted and analyzed the data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Shanghai Ninth People's Hospital, affiliated with the School of Medicine, Shanghai Jiao Tong University (approval no. 31600760; Shanghai, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Mantha S, Pillai S, Khayambashi P, Upadhyay A, Zhang Y, Tao O, Pham HM and Tran SD: Smart hydrogels in tissue engineering and regenerative medicine. Materials (Basel). 12(pii: E3323)2019.PubMed/NCBI View Article : Google Scholar

2 

Costa-Almeida R, Calejo I and Gomes ME: Mesenchymal stem cells empowering tendon regenerative therapies. Int J Mol Sci. 2(pii: E3002)2019.PubMed/NCBI View Article : Google Scholar

3 

Crupi A, Costa A, Tarnok A, Melzer S and Teodori L: Inflammation in tissue engineering: The Janus between engraftment and rejection. Eur J Immunol. 45:3222–3236. 2015.PubMed/NCBI View Article : Google Scholar

4 

Przekora A: The summary of the most important cell-biomaterial interactions that need to be considered during in vitro biocompatibility testing of bone scaffolds for tissue engineering applications. Mater Sci Eng C Mater Biol Appl. 97:1036–1051. 2019.PubMed/NCBI View Article : Google Scholar

5 

Liu C and Sun J: Potential application of hydrolyzed fish collagen for inducing the multidirectional differentiation of rat bone marrow mesenchymal stem cells. Biomacromolecules. 15:436–443. 2014.PubMed/NCBI View Article : Google Scholar

6 

Blanco M, Vázquez JA, Pérez-Martín RI and Sotelo CG: Hydrolysates of fish skin collagen: An opportunity for valorizing fish industry byproducts. Mar Drugs. 15(pii: E131)2017.PubMed/NCBI View Article : Google Scholar

7 

Liu C and Sun J: Hydrolyzed tilapia fish collagen induces osteogenic differentiation of human periodontal ligament cells. Biomed Mater. 10(065020)2015.PubMed/NCBI View Article : Google Scholar

8 

Raabe O, Reich C, Wenisch S, Hild A, Burg-Roderfeld M, Siebert HC and Arnhold S: Hydrolyzed fish collagen induced chondrogenic differentiation of equine adipose tissue-derived stromal cells. Histochem Cell Biol. 134:545–554. 2010.PubMed/NCBI View Article : Google Scholar

9 

Liu C, Liu X, Xue Y, Ding TT and Sun J: Hydrolyzed tilapia fish collagen modulates the biological behavior of macrophages under inflammatory conditions. RSC Adv. 5:30727–30736. 2015.

10 

Rackham CL and Jones PM: Potential of mesenchymal stromal cells for improving islet transplantation outcomes. Curr Opin Pharmacol. 43:34–39. 2018.PubMed/NCBI View Article : Google Scholar

11 

Mukherjee S, Darzi S, Paul K, Werkmeister JA and Gargett CE: Mesenchymal stem cell-based bioengineered constructs: Foreign body response, cross-talk with macrophages and impact of biomaterial design strategies for pelvic floor disorders. Interface Focus. 9(20180089)2019.PubMed/NCBI View Article : Google Scholar

12 

Yang M and Liu L: MHC II gene knockout in tissue engineering may prevent immune rejection of transplants. Med Hypotheses. 70:798–801. 2008.PubMed/NCBI View Article : Google Scholar

13 

Weston LE, Geczy AF and Briscoe H: Production of IL-10 by alloreactive sibling donor cells and its influence on the development of acute GVHD. Bone Marrow Transplant. 37:207–212. 2006.PubMed/NCBI View Article : Google Scholar

14 

Sica A and Mantovani A: Macrophage plasticity and polarization: In vivo veritas. J Clin Invest. 122:787–795. 2012.PubMed/NCBI View Article : Google Scholar

15 

Ben-Mordechai T, Palevski D, Glucksam-Galnoy Y, Elron-Gross I, Margalit R and Leor J: Targeting macrophage subsets for infarct repair. J Cardiovasc Pharmacol Ther. 20:36–51. 2015.PubMed/NCBI View Article : Google Scholar

16 

Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT and Sahebkar A: Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 233:6425–6440. 2018.PubMed/NCBI View Article : Google Scholar

17 

Bernardo ME and Fibbe WE: Mesenchymal stromal cells and hematopoietic stem cell transplantation. Immunol Lett. 168:215–221. 2015.PubMed/NCBI View Article : Google Scholar

18 

Deng W, Chen W, Zhang Z, Huang S, Kong W, Sun Y, Tang X, Yao G, Feng X, Chen W and Sun L: Mesenchymal stem cells promote CD206 expression and phagocytic activity of macrophages through IL-6 in systemic lupus erythematosus. Clin Immunol. 161:209–216. 2015.PubMed/NCBI View Article : Google Scholar

19 

Abumaree MH, Al Jumah MA, Kalionis B, Jawdat D, Al Khaldi A, Abomaray FM, Fatani AS, Chamley LW and Knawy BA: 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 Rep. 9:620–641. 2013.PubMed/NCBI View Article : Google Scholar

20 

Kim J and Hematti P: Mesenchymal stem cell-educated macrophages: A novel type of alternatively activated macrophages. Exp Hematol. 37:1445–1453. 2009.PubMed/NCBI View Article : Google Scholar

21 

Ripoll CB, Flaat M, Klopf-Eiermann J, Fisher-Perkins JM, Trygg CB, Scruggs BA, McCants ML, Leonard HP, Lin AF, Zhang S, et al: Mesenchymal lineage stem cells have pronounced anti-inflammatory effects in the twitcher mouse model of Krabbe's disease. Stem Cells. 29:67–77. 2011.PubMed/NCBI View Article : Google Scholar

22 

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.PubMed/NCBI View Article : Google Scholar

23 

Polymeri A, Giannobile WV and Kaigler D: Bone marrow stromal stem cells in tissue engineering and regenerative medicine. Horm Metab Res. 48:700–713. 2016.PubMed/NCBI View Article : Google Scholar

24 

Wallace CS and Truskey GA: Direct-contact co-culture between smooth muscle and endothelial cells inhibits TNF-alpha-mediated endothelial cell activation. Am J Physiol Heart Circ Physiol. 299:H338–H346. 2010.PubMed/NCBI View Article : Google Scholar

25 

Livak JK and Schmittgen TD: Analysis of relative gene expression data using quantitative PCR and the 2-ΔΔCt method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

26 

Shujia J, Haider HK, Idris NM, Lu G and Ashraf M: Stable therapeutic effects of mesenchymal stem cell-based multiple gene delivery for cardiac repair. Cardiovasc Res. 77:525–533. 2008.PubMed/NCBI View Article : Google Scholar

27 

Secunda R, Vennila R, Mohanashankar AM, Rajasundari M, Jeswanth S and Surendran R: Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: A comparative study. Cytotechnology. 67:793–807. 2015.PubMed/NCBI View Article : Google Scholar

28 

Climaco-Arvizu S, Domínguez-Acosta O, Cabañas-Cortés MA, Rodríguez-Sosa M, Gonzalez FJ, Vega L and Elizondo G: Aryl hydrocarbon receptor influences nitric oxide and arginine production and alters M1/M2 macrophage polarization. Life Sci. 155:76–84. 2016.PubMed/NCBI View Article : Google Scholar

29 

Arndt L, Dokas J, Gericke M, Kutzner CE, Müller S, Jeromin F, Thiery J and Burkhardt R: Tribbles homolog 1 deficiency modulates function and polarization of murine bone marrow-derived macrophages. J Biol Chem. 293:11527–11536. 2018.PubMed/NCBI View Article : Google Scholar

30 

Michelo CM, Fasse E, van Cranenbroek B, Linda K, van der Meer A, Abdelrazik H and Joosten I: Added effects of dexamethasone and mesenchymal stem cells on early Natural Killer cell activation. Transpl Immunol. 37:1–9. 2016.PubMed/NCBI View Article : Google Scholar

31 

Albu DI, Wang Z, Huang KC, Wu J, Twine N, Leacu S, Ingersoll C, Parent L, Lee W, Liu D, et al: EP4 Antagonism by E7046 diminishes Myeloid immunosuppression and synergizes with Treg-reducing IL-2-Diphtheria toxin fusion protein in restoring anti-tumor immunity. Oncoimmunology. 6(e1338239)2017.PubMed/NCBI View Article : Google Scholar

32 

Mattes B and Scholpp S: Emerging role of contact-mediated cell communication in tissue development and diseases. Histochem Cell Biol. 150:431–442. 2018.PubMed/NCBI View Article : Google Scholar

33 

Rahimzadeh A, Mirakabad FS, Movassaghpour A, Shamsasenjan K, Kariminekoo S, Talebi M, Shekari A, Zeighamian V, Ghalhar MG and Akbarzadeh A: Biotechnological and biomedical applications of mesenchymal stem cells as a therapeutic system. Artif Cells Nanomed Biotechnol. 44:559–570. 2016.PubMed/NCBI View Article : Google Scholar

34 

Liu C and Sun J: Osteogenically differentiated mesenchymal stem cells induced by hydrolyzed fish collagen maintain their immunomodulatory effects. Life Sci. 238(116970)2019.PubMed/NCBI View Article : Google Scholar

35 

Abdelrazik H, Spaggiari GM, Chiossone L and Moretta L: Mesenchymal stem cells expanded in human platelet lysate display a decreased inhibitory capacity on T- and NK-cell proliferation and function. Eur J Immunol. 41:3281–3290. 2011.PubMed/NCBI View Article : Google Scholar

36 

Duffy MM, Ritter T, Ceredig R and Griffin MD: Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2(34)2011.PubMed/NCBI View Article : Google Scholar

37 

Mills CD: M1 and M2 Macrophages: Oracles of health and disease. Crit Rev Immunol. 32:463–488. 2012.PubMed/NCBI View Article : Google Scholar

38 

Varela P, Sartori S, Viebahn R, Salber J and Ciardelli G: Macrophage immunomodulation: An indispensable tool to evaluate the performance of wound dressing biomaterials. J Appl Biomater Funct Mater. 17(2280800019830355)2019.PubMed/NCBI View Article : Google Scholar

39 

Mueller CK and Schultze-Mosgau S: Histomorphometric analysis of the phenotypical differentiation of recruited macrophages following subcutaneous implantation of an allogenous acellular dermal matrix. Int J Oral Maxillofac Surg. 40:401–407. 2011.PubMed/NCBI View Article : Google Scholar

40 

Dinarello CA: Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 27:519–550. 2009.PubMed/NCBI View Article : Google Scholar

41 

Nishimoto N: Interleukin-6 as a therapeutic target in candidate inflammatory diseases. Clin Pharmacol Ther. 87:483–487. 2010.PubMed/NCBI View Article : Google Scholar

42 

Batlle E and Massagué J: Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity. 50:924–940. 2019.PubMed/NCBI View Article : Google Scholar

43 

Gorelik L and Flavell RA: Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med. 7:1118–1122. 2001.PubMed/NCBI View Article : Google Scholar

44 

Steen EH, Wang X, Balaji S, Butte MJ, Bollyky PL and Keswani SG: the role of the anti-inflammatory cytokine interleukin-10 in tissue fibrosis. Adv Wound Care (New Rochelle). 9:184–198. 2020.PubMed/NCBI View Article : Google Scholar

45 

Mittal SK and Roche PA: Suppression of antigen presentation by IL-10. Curr Opin Immunol. 34:22–27. 2015.PubMed/NCBI View Article : Google Scholar

46 

Ferrante CJ, Pinhal-Enfield G, Elson G, Cronstein BN, Hasko G, Outram S and Leibovich SJ: The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Rα) signaling. Inflammation. 36:921–931. 2013.PubMed/NCBI View Article : Google Scholar

47 

Boehler RM, Kuo R, Shin S, Goodman AG, Pilecki MA, Gower RM, Leonard JN and Shea LD: Lentivirus delivery of IL-10 to promote and sustain macrophage polarization towards an anti-inflammatory phenotype. Biotechnol Bioeng. 111:1210–1221. 2014.PubMed/NCBI View Article : Google Scholar

48 

Ylöstalo JH, Bartosh TJ, Coble K and Prockop DJ: Human mesenchymal stem/stromal cells cultured as spheroids are self-activated to produce prostaglandin E2 that directs stimulated macrophages into an anti-inflammatory phenotype. Stem Cells. 30:2283–2296. 2012.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu C and Liu C: Modulation of the secretion of mesenchymal stem cell immunoregulatory factors by hydrolyzed fish collagen. Exp Ther Med 20: 375-384, 2020
APA
Liu, C., & Liu, C. (2020). Modulation of the secretion of mesenchymal stem cell immunoregulatory factors by hydrolyzed fish collagen. Experimental and Therapeutic Medicine, 20, 375-384. https://doi.org/10.3892/etm.2020.8674
MLA
Liu, C., Sun, J."Modulation of the secretion of mesenchymal stem cell immunoregulatory factors by hydrolyzed fish collagen". Experimental and Therapeutic Medicine 20.1 (2020): 375-384.
Chicago
Liu, C., Sun, J."Modulation of the secretion of mesenchymal stem cell immunoregulatory factors by hydrolyzed fish collagen". Experimental and Therapeutic Medicine 20, no. 1 (2020): 375-384. https://doi.org/10.3892/etm.2020.8674