Semaphorin 3A enhances osteogenesis of MG63 cells through interaction with Schwann cells in vitro

  • Authors:
    • Hongqiang Yu
    • Tingting Pei
    • Jingyi Ren
    • Ye Ding
    • Anqian Wu
    • Yanmin Zhou
  • View Affiliations

  • Published online on: February 22, 2018     https://doi.org/10.3892/mmr.2018.8628
  • Pages: 6084-6092
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bone remodeling is under the control of various signals and systems in the body, including the nervous system. Semaphorin (Sema) 3A is a chemorepellent protein which regulates bone mass. Schwann cells, having a pivotal role following nerve injury, interact with Sema3A under numerous circumstances. The present study established a co‑culture system of MG63 and Schwann cells to investigate the role of the interaction between Sema3A and Schwann cells in osteogenesis. The results from the alkaline phosphatase assay, calcium nodule staining and the analysis of the osteogenic gene expression revealed that Sema3A inhibits osteogenic differentiation of MG63 cells in single‑cell culture and promotes osteogenic differentiation of MG63 cells in co‑culture with Schwann cells, in a concentration‑dependent manner. These findings suggest that the presence of Schwann cells induces Sema3A‑associated osteogenic differentiation in bone cells, and also reveals the pivotal role of Sema3A as a regulator in the skeletal and nervous systems, thus contributing to a better understanding of the interaction between these systems.

Introduction

Bone remodeling is strongly regulated by hormones, cytokines and other cellular interactions that affect communication between osteoblasts and osteoclasts. Any disruption in this network may result in abnormal bone mass, including osteoporosis (1).

It has previously been demonstrated that the nervous system is an important regulator of bone formation. Due to the thick innervation in bone tissue, chemical messengers are transduced to bone and periosteum through the sensory and sympathetic nerve fibers (24). In addition, catecholaminergic positive axons may be visualized near osteoblasts in vivo and damaged or missing peripheral nerve fibers may result in abnormal bone formation (5,6). Furthermore, the existence of β-adrenergic receptors in osteoblasts and osteoclasts verifies the role of the central nervous system in the regulation of bone formation (7). However, the molecular mechanisms through which neurons and nerve fibers reach their targets and function during osteogenesis are still poorly understood.

In the central nervous system, several protein families involved in wiring, location, and migration of axons, affecting the length and branching of dendrites were previously reported. One of the most prevalent neural signaling molecular groups are the Semaphorins, a verified set of neuro-immune molecules, which exhibit a predominant role in cardiac and skeletal development, epithelial morphogenesis, angiogenesis and tumor regression (812). Semaphorin (Sema) 3A, the first protein discovered in this large protein family, is a chemorepellent for the nervous system that serves to induce the retraction and collapse of the structure of axonal growth cone, and to affect fiber plasticity in adults (1315). Sema3A completes its biological function via binding to a receptor complex encompassing ligand-binding component Neuropilin (Nrp) 1 and class A Plexins (Plx) a 1, 2, 3 and 4, the latter of which is necessary for proper signal transduction (16). Previously, Sema3A was reported to have a significant role in bone remodeling (1719). In a previous study, Sema3A-deficient mice and neuro-specific Sema3A-deficient mice were generated and both exhibited low bone mass, low expression of sensory-nerve markers and their positive nerve fibers, and defective innervations. Conversely, osteoblast-specific Sema3A-deficient mice and wild-type mice did not exhibit any of these malfunctions or abnormalities (20). These results suggested that the absence of Sema3A in osteoblasts was not the only factor resulting in bone defects and that Sema3A modulated sensory nerve innervation during bone remodeling. However, the specific role of Sema3A through which bone formation is affected is still unclear.

Previous studies have exposed the key role of Schwann cells in repair and reconstruction following peripheral nerve injury. Injury-induced Schwann cells undergo a series of alterations including downregulation of myelin genes, upregulation of trophic factors and cytokines, activation of myelin autophagy and invasion of macrophages. Altogether, these alterations lead to increased regeneration of axons, remyelination of nerve fibers, avoidance of tissue loss and improved function (2123). Furthermore, Schwann cell migration is significantly impaired following the loss of Nrp1 (24) and binding between Sema3A and Nrp1 has been described as a key interaction in bone cell differentiation (18,19). It was therefore hypothesized that during Sema3A-associated osteogenesis, neural cells, including Schwann cells, may exhibit a pivotal role by interacting with Sema3A and osteoblastic cells.

The present study investigated the role of neural cells in bone remodeling. Sema3A was added to a single cell culture of MG63, and to a co-culture of MG63 and Schwann cells. Osteogenic differentiation was assessed to determine if Sema3A osteogenic induction is influenced by the addition of Schwann cells.

Materials and methods

Materials

Recombinant human Sema3A protein was purchased from Sino Biological Inc. (Beijing, China). The human osteoblast cell line MG63 and the human immortalized Schwann cell line sNF96.2 (SCs) were provided by the School of Stomatology of Jilin University (Changchun, China). Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), and penicillin/streptomycin were purchased from Invitrogen; Thermo Fisher Scientific, Inc., (Waltham, MA, USA). Trypsin and Dulbecco's phosphate buffered saline (PBS) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Cell Counting Kit-8 (CCK8) was purchased from 7Sea Pharmatech (Shanghai, China). In Situ Cell Apoptosis Detection kit (AP kit) was purchased from Boster Biological Technology (Pleasanton, CA, USA). Total RNA was extracted using the TRIzol® method (Invitrogen; Thermo Fisher Scientific, Inc.). All absorbance represented by optical density (OD) value was measured using Synergy HT spectrophotometer (BioTek Instruments, Inc., Winooski, VT, USA).

Single and co-cell culture systems

The MG63 and SCs were cultured in DMEM supplemented with 10% FBS, 1% penicillin, and 1% streptomycin, according to previously reported culturing conditions (25,26). The medium was replaced every 3 days. Cells were trypsinized and centrifuged at 183 × g, 37°C for 5 min when the confluence reached 80%. Following centrifugation, each cell was re-suspended. A total of 500 cells were seeded in each well of 96-well plates at various MG63 to SCs proportions (1:0, 1:1, 1:2, 2:1 and 0:1) to establish single and co-culture systems. Cells were cultured at 37°C in a humidified atmosphere containing 5% CO2.

Cell proliferation and apoptosis assay

MG63 and SCs single-cell cultures and co-cultures were seeded onto 96-well plates and cultured for 5 days to test their level of proliferation. At days 1, 3 and 5, 10 µl of CCK8 reagent was added to each well. Cells were incubated at 37°C for 40 min for interaction with the reagent. Then the plate was examined by spectrophotometer at a wavelength of 450 nm to assess OD. The same test was conducted on the two single-cell cultures and the co-culture system of MG63 to SCs at 1:1 (1:1 co-culture), with addition of Sema3A to investigate if Sema3A affected proliferation. Briefly, Sema3A (dissolved in PBS) was added to each well of the 96-well plates to reach final concentration of 25, 50 and 100 ng/ml. At days 1, 3 and 5, cell proliferation was measured following CCK8 treatment by spectrophotometer (450 nm). Controls (0 ng/ml Sema3A) for both single-cell cultures and 1:1 co-culture, were created using the same volume of PBS. The apoptosis assay was carried out to investigate apoptosis in the 1:1 co-culture treated with Sema3A using 6-well plates at days 1, 3 and 5 by AP kit according to the manufacturer's protocol. The principle of this kit is that cells in apoptosis produce DNA breakpoints with 3′-OH terminals. Terminal Deoxynucleotidyl Transferase (TdT) tags digoxigenin (Dig)-marked dUTP to 3′-OH terminals of broken DNAs. Then by reaction with the anti-Dig-biotin and streptavidin-biotin complex/alkaline phosphatase (SABC/AP), broken DNA will become colored following the addition of a chromogenic substrate. Briefly, cells were first fixed in 4% paraformaldehyde at room temperature for 30 min, then went through reactions with TdT/Dig-dUTP, anti-Dig-biotin and SABC/AP respectively, finally were stained by 20X 5-bromo-4-chloro-3-indolyl phosphate/nitroblue tetrazolium at room temperature for 20 min and washed by 0.01 M Tris buffered saline adequately to remove the excess dye. Under microscope, nuclei of apoptotic cells were stained dark blue. For each group, three individual visual fields of 500 cells each were observed and the total amount of dead cells were counted.

Cytotoxicity assay

To assess the cytotoxicity of Sema3A, Sema3A was added to the culture medium to reach the final concentrations of 25, 50 and 100 ng/ml. Medium with the same volume of PBS was used as the control group (0 ng/ml Sema3A). 1:1 co-culture was maintained in each medium for 6, 12 and 24 h and then tested by CCK8 at each timepoint. OD values were measured by spectrophotometer (450 nm) following incubation for 40 min.

Expression of Sema3A receptors

Gene expression of Sema3A receptors in MG63 and SCs was analyzed using reverse transcription-semi quantitative polymerase chain reaction (RT-sqPCR). Total RNA was extracted from each cell group using the TRIzol method according to manufacturer's protocol. Total RNA density was measured using NanoDrop 2000c Spectrophotometer (Thermo Fisher Scientific, Inc.). Subsequently, complementary DNA (cDNA) was synthesized from 1 µg total RNA using the PrimeScript RT kit (Takara Bio, Inc., Otsu, Japan) following the manufacturer's protocol. RT-sqPCR amplification was carried out on cDNA using the Sapphire Amp Fast PCR Master Mix (Takara Bio, Inc.). Synthesis and amplification were conducted in an Alpha Thermal Cycler (Bibby Scientific, Ltd., Staffordshire, UK). Primers for the house-keeping gene β-actin as well as Sema3A receptors (Nrp1, Nrp2, Plxa1, Plxa2, Plxa3, and Plxa4) were used for amplification. Sequences of these primers, as previously described (27), are listed in Table I. Each 50 µl PCR reaction system consisted of 25 µl SapphireAmp Fast PCR Master Mix, 1 µl 10 µM forward primer, 1 µl 10 µM reverse primer, 2 µl 50 ng/µl cDNA, and 21 µl dH2O. The PCR cycle reaction was performed under the following conditions: Polymerase activation step at 94°C for 1 min followed by 30 cycles (98°C for 5 sec, 55°C for 5 sec, 72°C for 10 sec), and final extension step at 72°C for 10 min. The PCR products were then loaded onto a 2% agarose gel (Invitrogen; Thermo Fisher Scientific, Inc.) containing 0.5 µg/ml ethidium bromide (Invitrogen; Thermo Fisher Scientific, Inc.). The size of the amplified bands was assessed against a 1,000 bp DNA ladder (Takara Bio, Inc.). Bands were visualized and captured using Molecular Imager Gel Doc XR+ (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and Image Lab (v4.0; Bio-Rad Laboratories, Inc.).

Table I.

Primer sequences used for RT-sqPCR and RT-qPCR.

Table I.

Primer sequences used for RT-sqPCR and RT-qPCR.

GeneForward sequenceReverse sequence
β-actin TGGCACCCAGCACAATGAA CTAAGTCATAGTCCGCCTAGAAGCA
Nrp1 CTCCTGTTGTGTCTTCAGG CCCGATGAGGATCGGATTC
Nrp2 CATGCACTATGACACCCCTG ATGGGTTCCATGCAGTTCTC
Plxa1 GACTTCCTGCTGACCCTGAG GACTTCAACCTGAAGCCAGC
Plxa2 GCTACAAGAGCTGGGTGGAG CTCTCGGCTTGAAGAACCAC
Plxa3 CAGCAGATCGACTACAAGAC GCCGTGTCAGGTAGATCTC
Plxa4 TGTCAGGGTGTCAACGAGAGC ATACACCTGCTCCTTGGTGG
RUNX2 CACTGGCGCTGCAACAAGA CATTCCGGAGCTCAGCAGAATAA
Osteocalcin (OCN) CCCAGGCGCTACCTGTATCAA GGTCAGCCAACTCGTCACAGTC

[i] Nrp, Neuropilin; Plx, Plexin; RUNX2, Runt-related transcription factor 2; OCN, Osteocalcin; RT-sqPCR, reverse transcription-semi quantitative polymerase chain reaction; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.

Osteogenic mRNA expression

The expression of osteogenic genes was analyzed by RT-qPCR. MG63 and 1:1 co-culture were divided into four groups with the addition of Sema3A or PBS to the final concentration of 0, 25, 50, 100 ng/ml. The medium was replaced every 2 days. At days 3 and 7, total RNA of cells was obtained and cDNA was synthetized using the aforementioned procedure. Then, primers for Runt-Related Transcription Factor 2 (RUNX2) and Osteocalcin (OCN) were used for RT-qPCR amplification using SYBR Premix EX Taq II RT-PCR kit (Takara Bio, Inc.) in Stratagene Mx3005P (Agilent Technologies, Inc., Santa Clara, CA, USA). The primer sequences used for RUNX2 and OCN amplification were as previously described (26) and listed in Table I. Briefly, each 25 µl RT-qPCR mixture consisted of 12.5 µl SYBR Fast qPCR Mix, 1 µl 10 µM forward and reverse primers, 0.5 µl Rox Reference Dye (50×), 2 µl 10 ng/µl cDNA and 8 µl dH2O. Each mixture was programmed to go through pre-denaturation at 95°C for 30 sec, then 40 cycles of 5 sec at 95°C followed by 30 sec at 60°C. β-actin was used as the reference gene in RT-qPCR analysis and 2−ΔΔCq method was applied to normalize and analyze the data as previously described (27).

Alkaline phosphatase (ALP) assay

The MG63 single culture and 1:1 co-culture under different concentrations of Sema3A were assessed by the ALP assay. At days 7 and 14, ALP activity was measured using an Alkaline phosphatase assay kit (Beyotime Institute of Biotechnology, Shanghai, China) according to manufacturer's protocol. Briefly, cells were washed with PBS three times and then lysed using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology). Cells were then centrifuged at 183 × g, 37°C for 5 min. For the ALP assay, 50 µl supernatant and 50 µl reaction reagent was added to each well of the 96-well plate. A total of 100 µl stop buffer was finally added to each well following a 10 min incubation at 37°C. Then, OD value at a wavelength of 405 nm was measured.

Extracellular matrix (ECM) mineralization

The single culture MG63 and 1:1 co-culture was cultured 14 days for mineralization staining. Cells were washed twice with PBS and fixed in 95% ethanol for 15 min at room temperature. Then, the Alizarin Red solution (Sigma-Aldrich; Merck KGaA) was used to dye the calcium nodules at room temperature for 5 min. Cells were rinsed adequately with distilled water to remove excessive dye. Then, the dyed nodules were dissolved in 10% (w/v) cetylpyridinium chloride (Shanghai Yuan Ye Biotechnology Co., Ltd., Shanghai, China) solution and OD was measured at a wavelength of 620 nm.

Statistical analysis

For data analysis of cell proliferation, apoptosis, cytotoxicity assays, calcium nodule staining and alkaline phosphatase assay, one-way analysis of variance followed by post-hoc Tukey's test was applied using SPSS software, version 17.0 (SPSS, Inc., Chicago, IL, USA). Data are presented as the mean ± standard error of the mean. All statistical graphs were produced by graphing software OriginLab 8 (v8.0725; OriginLab, Northampton, MA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Establishment of cell culture systems

The three co-culture groups demonstrated different cell viabilities. The 1:1 co-culture was the group with the most increased cell proliferation at days 1, 3 and 5 (Fig. 1A). Other groups demonstrated inferior cell proliferation (P<0.05) compared with this group, at all time points examined. Cell proliferation of single MG63 and SCs were not increased compared with the 1:1 group. Hence, 1:1 of MG63 to SCs was the ratio selected for the follow-up experiments.

Effect of Sema3A on cell proliferation, apoptosis, and cytotoxicity

Cell proliferation, cell death, and cytotoxicity following Sema3A addition were assessed. There was no difference in cell proliferation among cell groups treated with different concentrations of Sema3A (0, 25, 50 and 100 ng/ml) at days 1, 3 and 5 (Fig. 1B). Similarly, the addition of Sema3A did not result in a significant alteration in cell proliferation of MG63 single culture during the testing period (Fig. 1C). The proliferation of SCs was also not affected by the addition of Sema3A at days 1 and 3. However, at day 5, OD value in 100 ng/ml Sema3A group was significantly decreased compared with 0, 25 and 50 ng/ml groups (P<0.05; Fig. 1D). This trend of reduction in the proliferation rate at day 5 may be observed with increasing concentrations of Sema3A (though not reaching statistical significance in other groups). For the apoptosis assay, there was no difference in levels of apoptosis in 1:1 co-culture, MG63 and SCs under different Sema3A concentrations at days 1, 3 and 5 (Fig. 1E). Similarly, there was no difference in levels of cytotoxicity in the 1:1 co-culture under different Sema3A concentrations (Fig. 1F).

Expression of Sema3A receptors

RT-sqPCR was performed to examine the expression of Sema3A receptors. Nrp1 was only expressed in MG63. Nrp2 expression was observed in SCs and was visible however weak in MG63. Gene expression of Plxa1 and Plxa2 was only detected in MG63. Plxa3 was the only Plexin receptor expressed in SCs (Fig. 2).

ALP assay and calcium nodule staining

An ALP assay was undertaken to assess the level of osteogenic differentiation. ALP activity in MG63 single culture indicated a consistent decline at days 7 and 14 (Fig. 3A). At each time point, only 50 and 100 ng/ml Sema3A groups suggested statistical difference from control group (0 ng/ml Sema3A; P<0.05). Furthermore, in MG63, ALP activity decreased gradually with increasing concentrations of Sema3A. Conversely, ALP activity in the 1:1 co-culture increased gradually as concentrations of Sema3A increased at days 7 and 14 (Fig. 3B). ALP assay results in 1:1 the co-culture revealed that all Sema3A groups (25, 50 and 100 ng/ml) were markedly increased compared with the control group (0 ng/ml;) at days 7 and 14, however, there was no difference among Sema3A treatment groups (Fig. 3B; P>0.05). The osteogenic differentiation was further investigated by calcium nodule staining. Differences in staining were not visually detectable (data not shown), however OD values indicated differences between groups. In MG63 single culture, OD value declined as concentration of Sema3A increased, and the lowest OD appeared at 100 ng/ml (Fig. 3C; P<0.05). No meaningful difference was observed between 25 and 50 ng/ml groups (Fig. 3C; P>0.05). In 1:1 co-culture, OD value demonstrated a steady increase as Sema3A concentration increased, with all groups statistically different compared with each other (P<0.05; Fig. 3C).

Expression of osteogenic genes

RT-qPCR was performed to detect alterations in expression levels of osteogenic genes under different concentrations of Sema3A. The pattern of alteration in mRNA expression of osteogenic genes was in accordance with the ALP activity and calcium nodule mineralization results. At day 3, RUNX2 and OCN expression in MG63 cells declined when Sema3A was added, with the lowest gene expression detected at 100 ng/ml Sema3A compared with control group (0 ng/ml). On the contrary, the gene expression levels of RUNX2 and OCN were increased in 1:1 co-culture with the highest expression detected at 100 ng/ml Sema3A (Fig. 4A and B). Similarly, at day 7, RUNX2 and OCN indicated the same pattern of expression as described at day 3 in both MG63 single culture and 1:1 co-culture. As concentration of Sema3A increased, MG63 demonstrated a decline in mRNA levels of both genes, whereas 1:1 co-culture demonstrated increased expression levels of the genes (Fig. 4C and D). All Sema3A treatment groups (25, 50 and 100 ng/ml) in MG63 and 1:1 co-culture were statistically different (P<0.05) from the control group (0 ng/ml) at every time point.

Discussion

The present study established a co-culture system of MG63 and Schwann cells to investigate the role of Sema3A and the mechanisms underlying bone formation. Although co-culture systems have been used in numerous studies, to the best of the author's knowledge, the present study was the first to use a co-culture system to dissect Sema3A function on osteogenesis. First, a co-culture system was generated and optimal cell proliferation determined at 1:1 of MG63:SCs. Cell proliferation in the 1:1 co-culture was consistently increased compared with MG63 and SCs single cultures. This is in accordance with a previous study, in which proliferation in a co-cultured system of Schwann cells and osteoblasts has been demonstrated to be consistently increased compared with single cell cultures (28). Then, the MG63 and SCs single cultures and 1:1 co-culture were treated with different concentrations of Sema3A (0, 25, 50 and 100 ng/ml) to observe any effect of Sema3A on cell proliferation. The results demonstrated that no statistical difference was present among groups in cell proliferation of MG63 and 1:1 co-culture, indicating no influence of Sema3A on cell growth. However, a decreased OD value in the 100 ng/ml Sema3A group of SCs was observed at day 5, suggesting that a high concentration of Sema3A inhibited proliferation of SCs. The apoptosis and cytotoxicity results for MG63 and SCs single culture and 1:1 co-culture also suggested that Sema3A at applied concentrations was non-cytotoxic and exerted no effect among treatment groups on apoptosis.

RT-sqPCR was then performed to investigate gene expression of Sema3A receptors in MG63 and SCs. Results verified that MG63 expressed Nrp1, 2 and Plxa1, a2 and a3. Nrp2 and Plxa3 expression was detected in SCs. Previous studies reported that Nrp1 is the primary binding ligand for Sema3A (29) and Nrp1-Sema3A binding is involved in regulation of the neural and immune system (24,3032). However, Nrp2, sharing 45% similarity in protein sequence with Nrp1 (33), has previously been implicated in Sema3A signaling. Glioma cell migration is regulated by Sema3A via Nrp2 (34), and Nrp2 improves Sema3A-dependent axonal targeting of the vomeronasal nerves in Nrp1 knockout mice (35). This contradicts with the previous view that Nrp1 bound preferentially with Sema3A and suggests a potential role for Nrp2 in Sema3A signaling. The present study observed low expression levels of Nrp2 in MG63 and high gene expression of Nrp2 in SCs. It was therefore proposed that Nrp2 of SCs may act as a principle receptor in Sema3A-mediated osteogenesis in the co-culture system. However, this still requires further investigation.

To dissect the role of Sema3A in osteogenesis and its interaction with neural cells, level of ALP, extracellular matrix mineralization and expression of osteogenic genes were assessed. Sema3A was first applied to MG63 single cell culture. However, contrary to previous findings on osteoblasts (36), ALP level, extracellular matrix mineralization and the expression of osteogenic genes suggested that Sema3A inhibited MG63 osteogenic differentiation compared with the control group (0 ng/ml Sema3A), with inhibitory levels rising with concentrations of Sema3A. Due to the lack of osteogenic medium, and low concentrations of Sema3A applied in the present study, 25 and 50 ng/ml Sema3A groups in calcium nodule staining and RUNX2 expression in RT-qPCR did not indicate a statistical difference. However, all Sema3A treatment groups, except the 25 ng/ml group of MG63 single culture in ALP assay, exhibited statistically significant differences compared with control group (0 ng/ml Sema3A) in all three assays. Furthermore, a trend of decline in osteogenic differentiation of MG63 was observed and verified in the present study. The authors present the following explanation to describe the decline in osteogenic expression in MG63 following Sema3A treatment. Firstly, MG63, alongside Saos-2 and U-2 OS cell lines, is a malignant bone tumor cell line acquired from osteosarcoma. Although several osteoblastic features are shared between these cell lines (37), cellular and molecular functions including intercellular communication may vary due to chromosome alterations (38). For instance, MG63 expresses collagen-II and -IX whereas no expression of these collagens is observed in human osteoblasts (39). The expression level of ALP in human osteoblasts is decreased compared with osteosarcoma cells and is dependent on the rate of confluence, which is different from MG63 (40). In addition, unlike osteoblasts, bone marrow mesenchymal stem cells do not express Nrp1 (41), whereas MG63 was verified to strongly express Nrp1 in the present study. Therefore, an altered chromosome expression of MG63 from human osteoblasts may exist, which may contribute to the deviated results of Sema3A function on osteogenesis from previous studies. Secondly, Sema3A has a complex role in cancer. Sema3A is downregulated in several cancers (42) and serves as an endogenous factor to inhibit tumor migration and progression (43). In addition, its binding with Nrp1 may inhibit cancer cell proliferation (44,45). In the present study, MG63 cells did not demonstrate any regression in proliferation under Sema3A. This may be due to the low concentrations applied. Sema3A may still exhibit an inhibitory role on the osteogenic differentiation of osteosarcoma-derived MG63, however to the best of the author's knowledge, no report has been discovered investigating the effect of Sema3A on osteosarcoma, and thus, its specific role in these cells requires further clarification. Furthermore, a previous study demonstrated that a low concentration of Sema3A may prevent Plexin receptors from performing signal transduction effectively due to insufficient binding with Sema3A, whereas other receptors including PlexinA3 may function properly at such concentrations (46). Therefore, instead of classic receptors including Nrp1, other ligands including Nrp2 and Plxa3, expression levels of which were verified in MG63 in the present study, may be involved in osteogenesis along with Sema3A under low concentrations. Sema3A may have engendered its osteogenic features on a dose-dependent basis due to receptor preference. However, the present study was rather limited in its ability to further illustrate the exact role of Sema3A in MG63 differentiation.

To explore the potential interaction between Sema3A and SCs in bone formation, the present study then added Sema3A at different concentrations to the 1:1 co-culture to test possible alterations in osteogenesis. The results indicated that ALP level, extracellular matrix mineralization, and osteogenic gene expression of 1:1 co-culture were gradually increased following addition of Sema3A, with the highest level observed at 100 ng/ml in all three tests, suggesting a dose-dependent pattern in skeletal function of Sema3A. However, 50 and 100 ng/ml groups in RT-qPCR did not reveal any statistical difference, indicating a more complicated role of concentration in Sema3A-induced osteogenesis. However, results revealed a positive role for SCs in bone formation. Schwann cells are of vital importance in nerve repair. Following peripheral nerve injury, class 3 Semaphorins including Sema3A are secreted by Schwann cells, and Nrp1/Nrp2 expression is upregulated in Schwann cells (47,48). This suggests the prominent involvement that Sema3A and SCs share in nerve regeneration. In the present study, the function of Sema3A in osteogenic expression in the 1:1 co-culture was completely reversed to that observed in the MG63 single culture. Firstly, according to a previous study (49), SCs excrete various numbers of neurotrophins including the brain-derived neurotrophic factor and the nerve growth factor (NGF) following injury. These neurotrophins, particularly NGF, have already been reported to have a role in osteoblastic survival (50), and they bind receptors on multiple non-neural cells including osteoblasts to inflict trophic features (51). Therefore, the addition of injury-associated Sema3A into the co-culture system may have promoted secretion of trophic factors in SCs, which then promoted MG63 differentiation. Secondly, since SCs were demonstrated to express Nrp2 and Plxa3 in the present study, the link between osteogenesis and Nrp2/Plxa3 was investigated. Verlinden et al (52) demonstrated that Nrp2 knock out mice have an insufficient number of osteoblasts, an increased level of osteoclasts, and low bone mass. Notably, it has previously been suggested that Nrp2, along with Plxa3, are receptors of Sema3A that only partially and subordinately participate in the signaling pathway in comparison to Nrp1 or Plxa1/2 (32,53). However, Nrp2 is required for SCs normal assembly into functional units for repair activities (54), and Plxa3 is necessary for nerve fibers to be precisely aimed at their targets (46). Therefore, whether this promotion in osteogenesis is in part or entirely due to the binding of Sema3A to Nrp2/Plxa3 or to other receptors, and semaphorins expressed and secreted by SCs to form functional complexes, including the bone-associated Semaphorin 6D-Plxa1[17] or the nerve-associated Semaphorin 3F-Nrp2 (52,55), remains unclear.

Following the loss of a tooth, a lot of periodontal exteroceptors and nerve fibers are lost. It is now well established that oral implants gain stability and function through osseointegration. However, numerous patients have reported having felt a sensory difference following implant restoration compared with their natural tooth, which may sometimes lead to occlusal overload. Klineberg et al (56) suggested the term ‘osseoperception’ in 2005 to describe these alterations in sensation. Neurophysiological and psychological explanations have been proposed recently, however how oral sensations may be re-transduced, or nerves be regenerated, is still unclear. In the present study, Sema3A promoted osteogenesis of MG63 via interaction with SCs, suggesting the potential of Sema3A in both the bone and nervous system, which may be applied in periodontal rehabilitation, helping to restore not only masticatory however also sensory, thus completing oral function.

In conclusion, the present study established a co-culture system to explore the potential role of Sema3A in osteogenesis. Results revealed that in vitro, low concentrations of Sema3A inhibited MG63 osteogenic expression. Conversely, by interaction with Schwann cells, osteogenic expression was elevated with increasing concentrations of Sema3A. The molecular mechanisms underlying the function of Sema3A in osteogenesis have not yet been fully elucidated, however assumptions have been put forward to sustain the hypothesis that Sema3A mediates bone cells via function of neural cells. Further investigations will help to better understand the exact role that the nervous system exhibit bone formation.

References

1 

Nakahama K: Cellular communications in bone homeostasis and repair. Cell Mol Life Sci. 67:4001–4009. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, Armstrong D, Ducy P and Karsenty G: Leptin regulates bone formation via the sympathetic nervous system. Cell. 111:305–317. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, Kondo H, Richards WG, Bannon TW, Noda M, et al: Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature. 434:514–520. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Serre CM, Farlay D, Delmas PD and Chenu C: Evidence for a dense and intimate innervation of the bone tissue, including glutamate-containing fibers. Bone. 25:623–629. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Lerner UH, Persson E and Lundberg P: Kinins and neuro-osteogenic factors. Principles Bone Biol. 2:1025–1057. 2008. View Article : Google Scholar

6 

Chenu C and Marenzana M: Sympathetic nervous system and bone remodeling. Joint Bone Spine. 72:481–483. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Togari A: Adrenergic regulation of bone metabolism: Possible involvement of sympathetic innervation of osteoblastic and osteoclastic cells. Microsc Res Tech. 58:77–84. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Toyofuku T, Zhang H, Kumanogoh A, Takegahara N, Yabuki M, Harada K, Hori M and Kikutani H: Guidance of myocardial patterning in cardiac development by Sema6D reverse signaling. Nat Cell Biol. 6:1204–1211. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Serini G, Valdembri D, Zanivan S, Morterra G, Burkhardt C, Caccavari F, Zammataro L, Primo L, Tamagnone L, Logan M, et al: Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature. 424:391–397. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Sierra JR, Corso S, Caione L, Cepero V, Conrotto P, Cignetti A, Piacibello W, Kumanogoh A, Kikutani H, Comoglio PM, et al: Tumor angiogenesis and progression are enhanced by Sema4D produced by tumor-associated macrophages. J Exp Med. 205:1673–1685. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Maione F, Molla F, Meda C, Latini R, Zentilin L, Giacca M, Seano G, Serini G, Bussolino F and Giraudo E: Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor growth and normalizes tumor vasculature in transgenic mouse models. J Clin Invest. 119:3356–3372. 2009.PubMed/NCBI

12 

Jongbloets BC and Pasterkamp RJ: Semaphorin signaling during development. Development. 141:3292–3297. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Luo Y, Raible D and Raper JA: Collapsin: A protein in brain that induces the collapse and paralysis of neuronal growth cones. Cell. 75:217–227. 1993. View Article : Google Scholar : PubMed/NCBI

14 

Tamagnone L and Comoglio PM: To move or not to move? Semaphorin signaling in cell migration. EBMO Rep. 5:356–361. 2004.

15 

Roth L, Koncina E, Satkauskas S, Crémel G, Aunis D and Bagnard D: The many faces of semaphorins: From development to pathology. Cell Mol Life Sci. 66:649–666. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Narazaki M and Tosato G: Ligand-induced internalization selects use of common receptor neuropilin-1 by VEGF-165 and Semaphorin 3A. Blood. 107:3892–3901. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Takegahara N, Takamatsu H, Toyofuku T, Tsujimura T, Okuno T, Yukawa K, Mizui M, Yamamoto M, Prasad DV, Suzuki K, et al: Plexin-A1 and its interaction with DAP12 in immune responses and bone homeostasis. Nat Cell Biol. 8:615–622. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Hayashi M, Nakashima T, Tanigushi M, Kodama T, Kumanogoh A and Takayanagi H: Osteoprotection by Semaphorin 3A. Nature. 485:69–74. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Negishi-Koga T, Shinohara M, Komatsu N, Bito H, Kodama T, Friedel RH and Takayanagi H: Suppression of bone formation by osteoblastic expression of semaphorin 4D. Nature Med. 17:1473–1480. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Fukuda T, Takeda S, Xu R, Ochi H, Sunamura S, Sato T, Shibata S, Yoshida Y, Gu Z, Kimura A, et al: Sema3A regulates bone-mass accrual through sensory innervations. Nature. 497:490–493. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Barrette B, Hébert MA, Filali M, Lafortune K, Vallières N, Gowing G, Julien JP and Lacroix S: Requirement of myeloid cells for axon regeneration. J Neurosci. 28:9363–9376. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Arthur-Farraj PJ, Latouche M, Wilton DK, Quintes S, Chabrol E, Banerjee A, Woodhoo A, Jenkins B, Rahman M, Turmaine M, et al: c-Jun reprograms Schwann cells of injured nerves to generate a repair cell essential for regeneration. Neuron. 75:633–647. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Jessen KR and Mirsky R: The repair Schwann cell and its function in regenerating nerves. J Physiol. 594:3521–3531. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Huttle RE and Huber AB: Cranial nerve fasciculation and Schwann cell migration are impaired after loss of Npn-1. Dev Biol. 359:230–241. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Tao Y: Isolation and culture of schwann cells. Methods Mol Biol. 1018:93–104. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Fang K, Song W, Wang L, Jia S, Wei H, Ren S, Xu X and Song Y: Immobilization of chitosan film containing Semaphorin 3A onto a microarc oxidized titanium implant surface via silane reaction to improve MG63 osteogenic differentiation. Int J Nanomedicine. 9:4649–4657. 2014.PubMed/NCBI

27 

Saad S, Dharmapatni AASSK, Crotti TN, Cantley MD, Algate K, Findlay DM, Atkins GJ and Haynes DR: Semaphorin-3a, neuropilin-1 and plexin-A1 in prosthetic-particle induced bone loss. Acta Biomater. 30:311–318. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Cai XX, Luo E and Yuan Q: Interaction between Schwann cells and osteoblasts in vitro. Int J Oral Sci. 2:74–81. 2010. View Article : Google Scholar : PubMed/NCBI

29 

He Z and Tessier-Lavigne M: Neuropilin is a receptor for the axonal chemorepellent Semaphorin III. Cell. 90:739–751. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Huettl RE, Soellner H, Bianchi E, Novitch BG and Huber AB: Npn-1 contributes to axon-axon interactions that differentially control sensory and motor innervation of the limb. PLoS Biol. 9:e10010202011. View Article : Google Scholar : PubMed/NCBI

31 

Bouvrée K, Brunet I, Del Toro R, Gordon E, Prahst C, Cristofaro B, Mathivet T, Xu Y, Soueid J, Fortuna V, et al: Semaphorin3A, neuropilin-1 and plexinA1 are required for lymphatic valve formation. Circ Res. 111:437–445. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M, Yagi T and Fujisawa H: A requirement for neuropilin-1 in embryonic vessel formation. Development. 126:4895–4902. 1999.PubMed/NCBI

33 

Cao Y, Hoeppner LH, Bach SEG, Guo Y, Wang E, Wu J, Cowley MJ, Chang DK, Waddell N, et al: Neuropilin-2 promotes extravasation and metastasis by interacting with endothelial α5 intergrin. Cancer Res. 73:4579–4590. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Nasarre C, Koncina E, Labourdette G, Cremel G, Roussel G, Aunis D and Bagnard D: Neuropilin-2 acts as a modulator of Sema3A-dependent glioma cell migration. Cell Adh Migr. 3:383–389. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Cariboni A, Davidson K, Rakic S, Maggi R, Parnavelas JG and Ruhrberg C: Defective gonadotropin-releasing hormone neuron migration in mice lacking SEMA3A signalling through NRP1 and NRP2: Implications for the aetiology of hypogonadotropic hypogonadism. Hum Mol Genet. 20:336–344. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Wan Q, Cho E, Yokota H and Na S: Rac1 and Cdc42 GTPases regulate shear stress-driven β-catenin signaling in osteoblasts. Biochem Biophys Res Commun. 433:502–207. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Declercq H, Van den Vreken N, De Maeyer E, Verbeeck R, Schacht E, De Ridder L and Cornelissen M: Isolation, proliferation and differentiation of osteoblastic cells to study cell/biomaterial interactions: Comparison of different isolation techniques and source. Biomaterials. 25:757–768. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Al-Romaih K, Bayani J, Vorobyova J, Karaskova J, Park PC, Zielenska M and Squire JA: Chromosomal instability in osteosarcoma and its association with centrosome abnormalities. Cancer Genet Cytogenet. 144:91–99. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Pautke C, Schieker M, Tischer T, Kolk A, Neth P, Mutschler W and Milz S: Characterization of osteosarcoma cell lines MG-63, Saos-2 and U-2 OS in comparison to human osteoblasts. Anticancer Res. 24:3743–3748. 2004.PubMed/NCBI

40 

Voegele TJ, Voegele-Kadletz M, Esposito V, Macfelda K, Oberndorfer U, Vecsei V and Schabus R: The effect of different isolation techniques on human osteoblast-like cell growth. Anticancer Res. 20:3575–3581. 2000.PubMed/NCBI

41 

Haixia D, Jingsong Z, Lei J, Hairong D, Jun W, Hang X and Weixian C: Gene expression of neuropilin-1 and its receptors, VEGF/Semaphorin 3A, in normal and cancer cells. Cell Biochem Biophys. 59:39–47. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Rehman M and Tamagnone L: Semaphorins in cancer: Biological mechanisms and therapeutic approaches. Semin Cell Dev Biol. 24:179–189. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Vacca A, Scavelli C, Serini G, Di Pietro G, Cirulli T, Merchionne F, Ribatti D, Bussolino F, Guidolin D, Piaggio G, et al: Loss of inhibitory Semaphorin 3A (SEMA3A) autocrine loops in bone marrow endothelial cells of patient with multiple myeloma. Blood. 108:1661–1667. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Catalano A, Caprari P, Rodilossi S, Betta P, Castellucci M, Casazza A, Tamagnone L and Procopio A: Cross-talk between vascular endothelial growth factor and semaphorin-3A pathway in the regulation of normal and malignant mesothelial cell proliferation. FASEB J. 18:358–360. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Herman JG and Meadows GG: Increased class 3 semaphorin expression modulates the invasive and adhesive properties of prostate cancer cells. Int J Oncol. 30:1231–1238. 2007.PubMed/NCBI

46 

Cheng HJ, Bagri A, Yaron A, Stein E, Pleasure SJ and Tessier-Lavigne M: Plexin-A3 mediates semaphorin signaling and regulates the development of hippocampal axonal projections. Neuron. 32:249–263. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Ara J, Bannerman P, Hahn A, Ramirez S and Pleasure D: Modulation of sciatic nerve expression of class 3 semaphorins by nerve injury. Neurochem Res. 29:1153–1159. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Scarlato M, Ara J, Bannerman P, Scherer S and Pleasure D: Induction of neuropilins-1 and −2 and their ligands, Sema3A, Sema3F and VEGF, during Wallerian degeneration in the peripheral nervous system. Exp Neurol. 183:489–498. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Alkhamarah BA, Hoshino N, Kawano Y, Harada F, Hanada K and Maeda T: The periodontal Ruffini endings in brain derived neurotrophic factor (BDNF) deficient mice. Arch Histol Cytol. 66:73–81. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Mogi M, Kondo A, Kinpara K and Togari A: Anti-apoptotic action of nerve growth factor in mouse osteoblastic cell line. Life Sci. 67:1197–1206. 2000. View Article : Google Scholar : PubMed/NCBI

51 

Mizuno N, Shiba H, Xu WP, Inui T, Fujita T, Kajiya M, Takeda K, Hasegawa N, Kawaguchi H and Kurihara H: Effect of neurotrophins on differentiation, calcification and proliferation in cultures of human pulp cells. Cell Biol Int. 31:1462–1469. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Verlinden L, Kriebitzsch C, Beullens I, Yan BK, Carmeliet G and Verstuyf A: Nrp2 deficiency leads to trabecular bone loss and is accompanied by enhanced osteoclast and reduced osteoblast numbers. Bone. 55:465–475. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Yuan L, Moyon D, Pardanaud L, Bréant C, Karkkainen MJ, Alitalo K and Eichmann A: Abnormal lymphatic vessel development in neuropilin 2 mutant mice. Development. 129:4797–4806. 2002.PubMed/NCBI

54 

Bannerman P, Ara J, Hahn A, Hong L, McCauley E, Friesen K and Pleasure D: Peripheral nerve regeneration is delayed in neuropilin 2-deficient mice. J Neurosci Res. 86:3163–3169. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Ara J, Bannerman P, Shaheen F and Pleasure DE: Schwann cell-autonomous role of neuropilin-2. J Neurosci Res. 79:468–475. 2005. View Article : Google Scholar : PubMed/NCBI

56 

Klineberg I, Calford MB, Dreher B, Henry P, Macefield V, Miles T, Rowe M, Sessle B and Trulsson M: A consensus statement on osseoperception. Clin Exp Pharmacol Physiol. 32:145–146. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 17 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu H, Pei T, Ren J, Ding Y, Wu A and Zhou Y: Semaphorin 3A enhances osteogenesis of MG63 cells through interaction with Schwann cells in vitro. Mol Med Rep 17: 6084-6092, 2018
APA
Yu, H., Pei, T., Ren, J., Ding, Y., Wu, A., & Zhou, Y. (2018). Semaphorin 3A enhances osteogenesis of MG63 cells through interaction with Schwann cells in vitro. Molecular Medicine Reports, 17, 6084-6092. https://doi.org/10.3892/mmr.2018.8628
MLA
Yu, H., Pei, T., Ren, J., Ding, Y., Wu, A., Zhou, Y."Semaphorin 3A enhances osteogenesis of MG63 cells through interaction with Schwann cells in vitro". Molecular Medicine Reports 17.4 (2018): 6084-6092.
Chicago
Yu, H., Pei, T., Ren, J., Ding, Y., Wu, A., Zhou, Y."Semaphorin 3A enhances osteogenesis of MG63 cells through interaction with Schwann cells in vitro". Molecular Medicine Reports 17, no. 4 (2018): 6084-6092. https://doi.org/10.3892/mmr.2018.8628