Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway

  • Authors:
    • Ruixin Zhen
    • Jianing Yang
    • Yu Wang
    • Yubo Li
    • Bin Chen
    • Youxin Song
    • Guiyun Ma
    • Bo Yang
  • View Affiliations

  • Published online on: February 6, 2018     https://doi.org/10.3892/mmr.2018.8559
  • Pages: 6045-6053
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bone regeneration is an important process associated with the treatment of osteonecrosis, which is caused by various factors. Hepatocyte growth factor (HGF) is an active biological factor that has multifunctional roles in cell biology, life sciences and clinical medicine. It has previously been suggested that bone morphogenetic protein (BMP)‑2 exerts beneficial roles in bone formation, repair and angiogenesis in the femoral head. The present study aimed to investigate the benefits and molecular mechanisms of HGF in bone regeneration. The viability of osteoblasts and osteoclasts were studied in vitro. In addition, the expression levels of tumor necrosis factor (TNF)‑α, monocyte chemotactic protein (MCP)‑1, interleukin (IL)‑1 and IL‑6 were detected in a mouse fracture model following treatment with HGF. The expression and activity of nuclear factor (NF)‑κB were also analyzed in osteocytes post‑treatment with HGF. Histological analysis was used to determine the therapeutic effects of HGF on mice with fractures. The migration and differentiation of osteoblasts and osteoclasts were investigated in HGF‑incubated cells. Furthermore, angiogenesis and BMP‑2 expression were analyzed in the mouse fracture model post‑treatment with HGF. The results indicated that HGF regulates the cell viability of osteoblasts and osteoclasts, and also balanced the ratio between osteoblasts and osteoclasts. In addition, HGF decreased the serum expression levels of TNF‑α, MCP‑1, IL‑1 and IL‑6 in experimental mice. The results of a mechanistic analysis demonstrated that HGF upregulated p65, IκB kinase‑β and IκBα expression in osteoblasts from experimental mice. In addition, the expression levels of vascular endothelial growth factor, BMP‑2 receptor, receptor activator of NF‑κB ligand and macrophage colony‑stimulating factor were upregulated by HGF, which may effectively promote blood vessel regeneration, and contribute to the formation and revascularization of tissue‑engineered bone. Furthermore, HGF promoted BMP‑2 expression and enhanced angiogenesis at the fracture location. These results suggested that HGF treatment may significantly promote bone regeneration in a mouse fracture model. In conclusion, these results indicated that HGF is involved in bone regeneration, angiogenesis and the balance between osteoblasts and osteoclasts, thus suggesting that HGF may be considered a potential agent for the treatment of fractures via the promotion of bone regeneration through regulation of the BMP‑2‑mediated NF‑κB signaling pathway.

Introduction

Bone regeneration is a biochemical process, which is required in response to the progression of cartilage wear and tear, damage and deformation caused by various types of joint disease (1). Previous studies have investigated the abilities of various proteins and cells to decrease inflammation associated with bone marrow, bone regeneration, vessel walls, adipose tissue, muscle, periosteum, tendons, peripheral circulation, umbilical cord blood, skin and dental tissues (24). In recent years, research has focused on the molecular mechanisms underlying bone regeneration and fracture repair (5,6). In addition, extracellular signaling molecules, including platelet-derived growth factor, fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) have been reported to promote fracture healing and bone regeneration (7). Furthermore, bone-healing processes, including vascularization, inflammatory inhibition and mesenchymal cell entry to the fracture site, have been reported to be mediated by extracellular signaling molecules (8). These findings indicate that targeted bone production and regeneration may contribute to the reduction of convalescence following treatment of fractures (1,9). The present study investigated the role of the extracellular signaling molecule hepatocyte growth factor (HGF) in inflammation, osteoblast and osteoclast balance, bone production and bone regeneration in a mouse fracture model. In addition, bone morphogenetic protein (BMP)-2 production and alterations in the nuclear factor (NF)-κB signaling pathway were detected in the mouse model.

HGF is produced by mesenchymal cells during organ injury and serves a crucial role in the process of bone regeneration (10). HGF is an active biological factor that exerts multifunctional effects on osteogenesis, and also serves a critical role in kidney development, acute injury and regeneration; HGF is activated by proteolytic cleavage at the site of injury resulting in formation of the biologically active HGF protein (11). A previous study reported that levels of HGF in the serum were correlated with quality of life in patients undergoing hemodialysis (12). In addition, biologically active HGF can suppress fibrosis; the molecular basis for HGF-mediated regression of renal fibrosis was elaborated in a previous report (13,14). Therefore, it was hypothesized that HGF may be regarded as a local acute phase protein that is beneficial for bone regeneration in various bone diseases. The results of the present study suggested that HGF promoted bone regeneration via regulation of BMP-2 expression in osteocytes in vitro and in vivo.

BMPs are members of the transforming growth factor-β superfamily, which regulate numerous cellular activities in bone regeneration (15). A previous study on BMP-2-releasing gelatin/β-tricalcium phosphate sponges revealed that BMP-2 may be considered a potential protein for the induction of bone regeneration in segmental bone defects (16). In addition, it has been reported that BMP-2 serves an important role in various forms of arthritis and disease activity. Grcevic et al (17) indicated that peripheral blood expression profiles of BMPs may be used as markers for arthritis, disease activity, therapeutic responsiveness and prognosis. Lories and Luyten also suggested that BMPs are beneficial for the repair of joint destruction and tissue responses that determine the outcome of chronic arthritis (18). Furthermore, BMP-2 is a member of the BMP family, which has been reported to possess high potential for bone formation, inflammatory inhibition in joints and synovial repair (19,20). In the present study, the in vivo effects and molecular mechanisms of BMP-2 were investigated in a mouse fracture model. Inflammation was also analyzed.

The present study aimed to investigate the association between bone tissue regeneration and inflammation in a mouse model of fracture. The results indicated that HGF treatment markedly promoted bone tissue regeneration and inhibited the expression of inflammatory factors. Notably, the present study demonstrated that HGF improves bone tissue regeneration via the BMP-2-mediated nuclear factor (NF)-κB signaling pathway, thus suggesting that HGF may be considered a promising agent for the treatment of patients with fractures.

Materials and methods

Ethics statement

The present study was conducted in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals at Chengde Medical College Affiliated Hospital (Chengde, China). The study was approved by the ethics committee of Chengde Medical College Affiliated Hospital. All surgeries and euthanasia were performed under intravenous sodium pentobarbital anesthesia (37 mg/kg).

Animal study

A total of 20 male C57BL/6J mice (6–8 weeks, 28–35 g) were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China). All animals were housed in a temperature-controlled facility at 23±1°C with a relative humidity of 50±5%, a 12 h light/dark cycle and unlimited access to food and water. Mice were subjected to an artificial fracture in the right paw as described previously (21). The mice were divided into two groups and were maintained under a normal schedule with free access to standard diet and water. On day 2 after model establishment, mice received treatment with HGF (10 mg/kg; Sigma Aldrich; Merck KGaA, Darmstadt, Germany) or the same volume of PBS (control) by intravenous injection. The body weights of the experimental mice were measured prior to each injection. Each mouse received 15 daily treatments. All mice were sacrificed on day 30 for histological analysis. Osteoblasts and osteoclasts were obtained from experimental mice and then isolated from one another as described previously (22,23) on day 30.

Cell culture

Osteocytes were obtained from mice with fractures prior to treatment as described previously (22,23) and then cultured in minimum essential media (MEM, Sigma-Aldrich; Merck KGaA) and supplemented with 10% fetal bovine serum (Sigma-Aldrich; Merck KGaA). Osteocytes were then treated with HGF (2 mg/ml) and cultured in a humidified incubator containing 5% CO2 for 24 h at 37°C.

MTT assay

Osteoblasts and osteoclasts were separately cultured in Dulbecco's Modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) and supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) for 24 h at 37°C. Osteoblasts and osteoclasts were then incubated with HGF (2 mg/ml) in a 96-well plate for 24 h at 37°C. A total of 20 µl MTT (5 mg/ml) in PBS solution was added to each well and cells were then incubated for a further 4 h at 37°C. Medium was then removed and 100 µl dimethyl sulfoxide was added to dissolve the formazan crystals. The viability of the cells was then determined using an ELISA reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA) at a wavelength of 450 nm.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from osteocytes using the RNA Easy Mini Extract kit (Sigma-Aldrich; Merck KGaA) according to the manufacturer's protocol. RNA was reversed transcribed using a PrimeScript RT Master Mix kit (Takara Bio, Inc., Otsu, Japan). Subsequently, cDNA (10 µg) was diluted 1/10 with distilled water and 10 µl was used for amplification. Specific primer sets for C-C motif chemokine ligand 2 (Ccl)2, Ccl5 and intercellular adhesion molecule 1 (Icam1) were conserved in the laboratory (Table I). RT-qPCR was performed using a qPCR system (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with SYBR Green Master Mix (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. A total of 45 amplification cycles were performed, including 94°C for 30 sec, denaturation at 96°C for 5 sec, primer annealing at 64°C for 5 sec, touchdown to 56°C for 15 sec and extension at 72°C for 10 sec. Relative mRNA expression levels were then determined using the 2−ΔΔCq method (24). The final results were presented as the fold of β-actin.

Table I.

Primers used for reverse transcription-quantitative polymerase chain reaction.

Table I.

Primers used for reverse transcription-quantitative polymerase chain reaction.

Target geneForward primerReverse primer
Ccl2 5′-AAGAAGCTGTAGTATTTGTCACCAAGCTCA-3′ 5′-CATCAGGTACGATCCAGGCT-3′
Ccl5 5′-GCAGCTGCATCCCTCACCGT-3′ 5′-GCAGCAGGGAGTGGTGTCCG-3′
Icam1 5′-GCTGTGCTTTGAGAACTGTG-3′ 5′-GTGAGGTCCTTGCCTACTTG-3′
β-actin 5′-GTGGGCGCCCAGGCACCA-3′ 5′-CTCCTTAATGTCACGCACGATTT-3′

[i] Ccl, C-C motif chemokine; Icam1, intercellular adhesion molecule 5.

Osteocyte migration assay

Osteocytes were obtained from mice with fractures, and osteoblasts and osteoclasts were isolated from mice that had undergone fracture prior to treatment as described in a previous study (25). Osteoblasts and osteoclasts were suspended at a density of 1×106 in 100 µl serum-free MEM for 12 h. The cells were then seeded into the upper chambers of a BD BioCoat Matrigel Invasion Chamber (BD Biosciences, Franklin Lakes, NJ, USA) and then incubated with HGF (2 mg/ml) for 12 h at 37°C. The migration assay was conducted according to the manufacturer's protocol.

Determination of NF-κB activity

Osteoblasts (1×106 cells) and osteoclasts (1×106 cells) were separately cultured in 6-well plates and incubated with either HGF (2 mg/ml) or PBS for 48 h. The efficacy of HGF on NF-κB activity was analyzed by determination of NF-κB-luciferase activity in cells. Briefly, cells were transfected with a pNF-κB-luciferase vector (Promega Corporation, Madison, WI, USA) for 48 h at 37°C using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to manufacturer's instructions. Luciferase activity in the cells was determined 72 h post-transfection at 37°C by using the Dual-Luciferase Reporter Assay System (Promega Corporation) following the manufacturer's protocol. Relative luciferase activity was determined by analyzing the firefly luciferase activity and normalizing it with the Renilla luciferase activity.

Cells differentiation

Osteoblasts and osteoclasts were obtained from mice with fractures prior to treatment and then separately cultured with MEM medium supplemented with 10% FBS. Cells were incubated with HGF (2 mg/ml) or the same volume of PBS (2 mg/ml) and then cultured for 72 h at 37°C. Analysis of tartrateresistant acid phosphatase activity was used to determine osteoblast and osteoclast differentiation.

Western blotting

Osteocytes were obtained from mice with fractures and were homogenized in lysis buffer containing protease inhibitor to perform protein extraction (Sigma-Aldrich; Merck KGaA), after which the cells were centrifuged at 6,000 × g at 4°C for 10 min. The supernatant was used for protein analysis. Protein concentration was determined using a BCA protein assay kit (Thermo Fisher Scientific, Inc.). Protein samples (10 µg) were separated using 12.5% SDS-PAGE, as described in a previous study (23), and then transferred to polyvinylidene fluoride (PVDF) membranes (EMD Millipore, Billerica, MA, USA). For western blotting, following blocking with 5% skimmed milk for 1 h at 37°C, primary antibodies: VEGF (cat. no. ab32152; 1:1,000; Abcam, Cambridge, UK), BMP-2 receptor [(R); cat. no. ab38560; 1:1,000; Abcam)], macrophage colony-stimulating factor (M-CSF; cat. no. ab9693; 1:1,000; Abcam), receptor activator of NF-κB ligand (RANKL; cat. no. ab216484; 1:1,000; Abcam) and β-actin (cat. no. ab8226; 1:1,000; Abcam) were then incubated with PVDF membranes for 12 h at 4°C. Subsequently, membranes were incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG mAb (cat. no. PV-6001; 1:5,000; OriGene Technologies, Inc., Beijing, China) for 24 h at 4°C. Following this, protein bands were visualized using Western Bright Enhanced Chemiluminescent HRP Substrate (Advansta, Inc., Menlo Park, CA, USA).

Small interfering (si)RNA transfections

Osteocytes (1×106 cells) were cultured to 80% confluence and were transfected with siRNA sequences (100 pM; Invitrogen; Thermo Fisher Scientific, Inc.) that targeted BMP-2: si-BMP-2 sense, 5′-GUGCUAUCUCGAUGCUGUATT-3′ and antisense, 5′-AAUACAGCAUCGAGAUAGCAC-3′; or scramble siRNA sense, 5′-GUGCUAUCUCGAUGCUGUA-3′ and antisense, 5′-UACAGCAUUCGAGAUAGCAC-3′. Transfection was performed using Lipofectamine® RNAi MAX (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Cells were cultured for 48 h at 37°C following transfection for further analysis.

Flow cytometry

On day 30, serum was obtained from experimental mice via centrifugation at 6,000 × g for 15 min at 4°C. Serum levels of lymphocytes, plasmacytes, neutrophils and monocytes in mice with fracture were analyzed by flow cytometry. All procedures were performed as described in a previous study (26). In addition, the ratios of apoptotic cells were determined using a Coulter EPICS XL Flow Cytometer (Beckman Coulter, Inc., Brea, CA, USA), and results were analyzed using Expo32-ADC v. 1.2B software (Beckman Coulter, Inc.).

ELISA

On day 30, to detect serum protein expression levels of inflammatory factors, mouse tumor necrosis factor (TNF)-α (cat. no. MTA00B; Bio-Rad Laboratories, Inc.), monocyte chemotactic protein (MCP)-1 (cat. no. CPCA00; Bio-Rad Laboratories, Inc.), interleukin (IL)-1 (cat. no. MLB00C; Bio-Rad Laboratories, Inc.) and IL-6 (cat. no. M6000B; Bio-Rad Laboratories, Inc.) ELISA kits were used. The experiments were conducted according to the manufacturer's protocols. Finally, absorbance of the samples was measured at 450 nm using an ELISA plate reader.

Histological analysis

To determine the therapeutic effects of HGF on a mouse model of fracture, the mice were sacrificed under pentobarbital anesthesia on day 30 and the bone tissues located in the fracture were separated and fixed in 10% formalin for 30 min at 37°C. The tissues were subsequently decalcified and embedded in paraffin for 2 h at 37°C. Paraffin-embedded bone tissues sections (4 µg) from experimental mice were stained with hematoxylin and eosin for 2 h at 37°C. Images were obtained using an inverted light microscope (Olympus Corporation, Tokyo, Japan).

Vascular density measurements and evaluation of bone resorption activity

Vascular density measurements and bone resorption activity were analyzed in mice following treatment with HGF, according to methods described in previous studies (27,28).

Statistical analysis

All data are presented as the mean ± standard deviation of triplicate experiments. All data were analyzed using SPSS Statistics 19.0 (IBM Corp., Armonk, NY, USA). Unpaired data were analyzed using Student's t-test P<0.05 was considered to indicate a statistically significant difference.

Results

In vitro effects of HGF treatment on migration and differentiation of osteocytes from a mouse model of fracture

The present study analyzed the effects of HGF on migration and differentiation of osteoblasts and osteoclasts from osteocytes obtained from mice with fracture. As shown in Fig. 1A, HGF promoted cell viability of osteoblasts and inhibited the cell viability of osteoclasts. A migration assay indicated that HGF treatment slightly enhanced migration of osteoblasts; however, no effects were detected on osteoclast migration (Fig. 1B). In addition, the results demonstrated that HGF promoted differentiation of osteoblasts and inhibited differentiation of osteoclasts at the fracture location (Fig. 1C). Furthermore, the balance of osteoblasts and osteoclasts (Ob/Oc) was improved in the fracture location (Fig. 1D). Taken together, these results suggested that HGF treatment may regulate migration and differentiation of osteoblasts and osteoclasts in a mouse model of fracture.

Effects of HGF treatment on inflammatory factors and biochemical analysis in a mouse model of fracture

A previous study indicated that aggravation of inflammatory responses can inhibit bone regeneration (29). Therefore, the present study detected inflammatory factors and biochemical indicators in a mouse model of fracture following treatment with HGF. As shown in Fig. 2A-D, the expression levels of TNF-α, MCP-1, IL-1 and IL-6 were downregulated in serum samples from HGF-treated experimental mice. Biochemical analysis indicated that the percentage of lymphocytes, plasmacytes, neutrophils and monocytes were also decreased in mice treated with HGF compared with PBS (Fig. 2E-H). These results suggested that HGF may be beneficial for the treatment of fractures in a mouse model via the downregulation of inflammatory responses.

Effects of HGF treatment on the expression levels of extracellular signaling molecules in a mouse model of fracture

After analyzing the alterations in inflammatory responses in a mouse model of fracture, the present study investigated the expression levels of extracellular signaling molecules. As presented in Fig. 3, the expression levels of VEGF, BMP-2R, RANKL and M-CSF were upregulated in the fracture location of HGF-treated mice. HGF-induced expression of extracellular signaling molecules may contribute to bone regeneration and bone healing in a mouse model of fracture. Taken together, these results suggested that HGF treatment increased the expression levels of extracellular signaling molecules in osteocytes in a mouse model of fracture.

Analysis of the mechanism underlying HGF-mediated NF-κB signaling in a mouse model of fracture

A previous study reported that NF-κB serves an essential role in bone regeneration via the regulation of numerous genes involved in cellular activity (30). Therefore, the present study analyzed the association between HGF and the NF-κB signaling pathway in a mouse model of fracture. As shown in Fig. 4A, treatment with HGF promoted NF-κB activity in osteocytes. In addition, p65, IKK-β and IκBα expression levels were upregulated in osteocytes from experimental mice treated with HGF (Fig. 4B). HGF treatment also increased the expression of the following anti-inflammatory genes, Ccl2, Ccl5 and Icam1, which are regulated by the NF-κB signaling pathway, as determined by RT-qPCR (Fig. 4C). Furthermore apoptosis of osteocytes was decreased in experimental mice treated with HGF (Fig. 4D). Taken together, these results indicated that HGF may regulate the physiological function of osteocytes via regulation of the NF-κB signaling pathway.

Analysis of HGF treatment on the expression levels of BMP-2 and revascularization-associated factors in a mouse model of fracture

The present study investigated the expression levels of BMP-2 and revascularization-associated factors in osteocytes from a mouse model of fracture. As shown in Fig. 5A, HGF upregulated BMP-2 expression in osteocytes from a mouse model of fracture. In addition, the expression levels of epidermal growth factor and FGF were increased in osteocytes from a mouse model of fracture (Fig. 5B). Inhibition of BMP-2 expression using siRNA (Si-BMP-2) decreased BMP-2 protein expression and inhibited NF-κB activity in osteocytes in vitro (Fig. 5C and D). Furthermore, the results indicated that Si-BMP-2 suppressed the expression levels of p65, IKK-β and IκBα in osteocytes in vitro (Fig. 5E). Taken together, these results suggested that HGF may enhance the NF-κB pathway via regulation of BMP-2 expression.

In vivo effects of HGF treatment on pathological alterations in a mouse model of fracture

The in vivo effects of HGF on a mouse model of fracture were also analyzed in the present study. As shown in Fig. 6A, HGF treatment increased body weight compared with in the PBS group. HGF also increased the bone resorption activity of mice (Fig. 6B). In addition, the vascular density of the HGF group was significantly increased compared with in the PBS group (Fig. 6C). The extent of bone regeneration and healing was also improved in HGF-treated fractured mice (Fig. 6D). These data suggested that HGF treatment may markedly improve bone resorption, neovascularization and bone regeneration, as determined by histological staining.

Discussion

Bone regeneration serves an essential role in fracture recovery, and is associated with bone resorption and bone-bonding ability (31,32). Previous studies have demonstrated that HGF is associated with cell regeneration and regulation of material-related bone formation (33,34). Although previous studies have indicated that HGF is constitutively produced by donor-derived bone marrow cells and can promote regeneration of pancreatic β-cells (35,36), the effects of HGF on bone regeneration have not been reported in previous studies. The present study investigated the effects, and potential molecular mechanisms, of HGF on bone regeneration in osteoblasts and osteoclasts in vitro and in fractured mice in vivo. The present study also studied the effects of HGF on inflammation in serum and apoptosis of osteocytes. The results indicated that HGF enhances the NF-κB pathway through the regulation of BMP-2 expression, which may markedly improve bone resorption, neovascularization and bone regeneration in a mouse model of fracture.

Theoretically, inflammatory cytokines are crucial regulatory factors in fracture progression that are mediated by various intracellular kinase signaling pathways, and regulate the local inflammatory response following acute tibial plateau fracture (37,38). Although a previous study reported inflammatory/necrosis biomarkers in fractured femurs treated with proximal femoral nail antirotation (39), the molecular mechanism has not been clearly elaborated in previous reports. In addition, IL-6 production modulated by HGF has been investigated in bone marrow-derived macrophages; the results revealed the role of HGF in inflammatory response-mediated diseases (40). Furthermore, overexpression of HGF in polycythemia vera attenuates the growth of clonal erythroblasts independently of V617F activating mutation of janus kinase 2 (41). Therefore, blocking inflammatory factors may interrupt the inflammatory process, thus breaking the vicious cycle of inflammation and promoting bone regeneration (42,43). In the present study, HGF exerted beneficial effects on the treatment of a mouse model of fracture by downregulation of inflammatory responses. In addition, HGF upregulated BMP-2 expression in a mouse model of fracture.

BMPs are members of the transforming growth factor-β superfamily, which regulate cellular metabolism, signaling pathways and numerous cell functions, including cell proliferation, migration, apoptosis, differentiation and adhesion (44,45). In recent years, reports have suggested that BMP-2 may be used to reconstruct segmental mandibular defects and effectively repair ischemic damage by inducing angiogenesis and osteogenesis, and by decreasing osteoclast bone resorption activity (46,47). In the present study, the association between HGF and BMP-2 was analyzed in a mouse model of fracture. Previous studies have reported that HGF contributes to fracture repair by upregulating the expression of BMPRs (48,49). It has also been suggested that BMP-2 promotes bone formation and osteoblastic differentiation by endochondral ossification (50,51). In addition, downregulation of BMPR expression is exhibited in mesenchymal cells from the joints of patients with rheumatoid arthritis (52). Furthermore, BMP-2 been clinically applied for spinal fusion procedures and it has been reported effectively regulate joint inflammation and damage (53,54). The results of the present study confirmed that HGF is an efficient drug for promotion of bone regeneration via BMP-2-induced NF-κB signaling.

A previous study indicated that the NF-κB signaling pathway has an essential role in bone regeneration and neovascularization via regulating the expression of VEGF, BMP-2, RANKL and M-CSF expression in fracture progression (55). Notably, HGF has been reported to preferentially stimulate NF-κB signaling, in order to protect renal proximal tubular epithelial cells against inflammation (43). In addition, BMP-2 exerts regulatory effects on apoptosis of chondrocytes, which serves a crucial role in the survival of chondrocytes (56). Furthermore, the NF-κB signaling pathway may stimulate BMP-2 gene expression in growth plate chondrocytes in vivo and in a chondrocyte cell line in vitro (57). In the present study, HGF was revealed to stimulate BMP-2-mediated NF-κB signaling, which may be responsible for the downregulation of the expression of TNF-α, IL-6 and IL-1 inflammatory factors. Outcomes have indicated that HGF-mediated NF-κB signaling may be attributed to BMP-2 upregulation, which may promote bone regeneration.

In conclusion, the results of the present study suggest that further investigation is required to determine the overall role of HGF in entire joint cytokine homeostasis, neovascularization and bone regeneration. The present findings indicated that HGF is beneficial for bone regeneration via increased expression of BMP-2, which leads to neovascularization and bone regeneration through regulation of the NF-κB signaling pathway. These preclinical data provided information that may be useful for the future treatment of patients with fractures.

References

1 

Furuya H, Tabata Y and Kaneko K: Bone regeneration for murine femur fracture by gelatin hydrogels incorporating basic fibroblast growth factor with different release profiles. Tissue Eng Part A. 20:1531–1541. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Wang X, Wang Y, Gou W, Lu Q, Peng J and Lu S: Role of mesenchymal stem cells in bone regeneration and fracture repair: A review. Int Orthop. 37:2491–2498. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Thorimbert V, König D, Marro J, Ruggiero F and Jaźwińska A: Bone morphogenetic protein signaling promotes morphogenesis of blood vessels, wound epidermis, and actinotrichia during fin regeneration in zebrafish. FASEB J. 29:4299–4312. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Perez RA, Kim JH, Buitrago JO, Wall IB and Kim HW: Novel therapeutic core-shell hydrogel scaffolds with sequential delivery of cobalt and bone morphogenetic protein-2 for synergistic bone regeneration. Acta Biomater. 23:295–308. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Rozen N, Lewinson D, Bick T, Meretyk S and Soudry M: Role of bone regeneration and turnover modulators in control of fracture. Crit Rev Eukaryot Gene Expr. 17:197–213. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Hutchison C, Pilote M and Roy S: The axolotl limb: A model for bone development, regeneration and fracture healing. Bone. 40:45–56. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Hankenson KD, Gagne K and Shaughnessy M: Extracellular signaling molecules to promote fracture healing and bone regeneration. Adv Drug Deliv Rev. 94:3–12. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Suzuki A, Uemura T and Nakamura H: Control of bone remodeling by nervous system. Neural involvement in fracture healing and bone regeneration. Clin Calcium. 20:1820–1827. 2010.(In Japanese).

9 

Tewari D, Khan MP, Sagar N, China SP, Singh AK, Kheruka SC, Barai S, Tewari MC, Nagar GK, Vishwakarma AL, et al: Ovariectomized rats with established osteopenia have diminished mesenchymal stem cells in the bone marrow and impaired homing, osteoinduction and bone regeneration at the fracture site. Stem Cell Rev. 11:309–321. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Ramezani A, Nägga K, Hansson O, Lönn J, Sjöwall J, Katoozian F, Mansouri S and Nayeri F: Hepatocyte growth factor in cerebrospinal fluid differentiates community-acquired or nosocomial septic meningitis from other causes of pleocytosis. Fluids Barriers CNS. 12:222015. View Article : Google Scholar : PubMed/NCBI

11 

Faletto DL, Kaplan DR, Halverson DO, Rosen EM and Vande Woude GF: Signal transduction in c-met mediated motogenesis. EXS. 65:107–130. 1993.PubMed/NCBI

12 

Baum E, Pawlaczyk K, Mackowiak B, Maćkowiak B, Sosinska P, Matecka M, Kolodziejczak B, Musielak M and Breborowicz A: Levels of hepatocyte growth factor in serum correlate with quality of life in hemodialysis patients. Int J Clin Exp Pathol. 8:13477–13482. 2015.PubMed/NCBI

13 

Mizuno S and Nakamura T: Molecular basis for HGF-mediated regression of renal fibrosis. Nihon Rinsho. 64 Suppl 2:S312–S321. 2006.

14 

Zhang SH, Wen KM, Wu W, Li WY and Zhao JN: Efficacy of HGF carried by ultrasound microbubble-cationic nano-liposomes complex for treating hepatic fibrosis in a bile duct ligation rat model, and its relationship with the diffusion-weighted MRI parameters. Clin Res Hepatol Gastroenterol. 37:602–607. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Singh K, Massel DH, Mayo BC, Bohl DD, Long WW and Modi KD: Bone morphogenetic proteins in lumbar arthrodesis: What is all the debate about? Commentary on an article by Daniel C. Beachler, PhD, MHS et al: ‘Bone morphogenetic protein use and cancer risk among patients undergoing lumbar arthrodesis: A case-cohort study using the SEER-medicare database’. J Bone Joint Surg Am. 98:e572016. View Article : Google Scholar : PubMed/NCBI

16 

Yamamoto M, Hokugo A, Takahashi Y, Nakano T, Hiraoka M and Tabata Y: Combination of BMP-2-releasing gelatin/β-TCP sponges with autologous bone marrow for bone regeneration of X-ray-irradiated rabbit ulnar defects. Biomaterials. 56:18–25. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Grcevic D, Jajic Z, Kovacic N, Lukic IK, Velagic V, Grubisic F, Ivcevic S and Marusic A: Peripheral blood expression profiles of bone morphogenetic proteins, tumor necrosis factor-superfamily molecules, and transcription factor Runx2 could be used as markers of the form of arthritis, disease activity, and therapeutic responsiveness. J Rheumatol. 37:246–256. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Lories RJ and Luyten FP: Bone morphogenetic proteins in destructive and remodeling arthritis. Arthritis Res Ther. 9:2072007. View Article : Google Scholar : PubMed/NCBI

19 

Postigo J, Iglesias M, Álvarez P, Jesús Augustin J, Buelta L, Merino J and Merino R: Bone morphogenetic protein and activin membrane-bound inhibitor, a transforming growth factor β rheostat that controls murine treg cell/Th17 cell differentiation and the development of autoimmune arthritis by reducing interleukin-2 signaling. Arthritis Rheumatol. 68:1551–1562. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Brescia AC, Simonds MM, McCahan SM, Fawcett PT and Rose CD: The role of transforming growth factor β signaling in fibroblast-like synoviocytes from patients with oligoarticular juvenile idiopathic arthritis: Dysregulation of transforming growth factor β signaling, including overexpression of bone morphogenetic protein 4, may lead to a chondrocyte phenotype and may contribute to bony hypertrophy. Arthritis Rheumatol. 66:1352–1362. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Holstein JH, Schmalenbach J, Herrmann M, Ölkü I, Garcia P, Histing T, Herrmann W, Menger MD, Pohlemann T and Claes L: Excess dietary methionine does not affect fracture healing in mice. Med Sci Monit. 18:BR469–BR474. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Wan Q, Schoenmaker T, Jansen ID, Bian Z, de Vries TJ and Everts V: Osteoblasts of calvaria induce higher numbers of osteoclasts than osteoblasts from long bone. Bone. 86:10–21. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Aino M, Nishida E, Fujieda Y, et al: Isolation and characterization of the human immature osteoblast culture system from the alveolar bones of aged donors for bone regeneration therapy. Expert Opin Biol Ther. 14:1731–1744. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Wai-Hoe L, Wing-Seng L, Ismail Z and Lay-Harn G: SDS-PAGE-based quantitative assay for screening of kidney stone disease. Biol Proced Online. 11:145–160. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Bajnok A, Kaposi A, Kovács L, Vásárhelyi B, Balog A and Toldi G: Analysis by flow cytometry of calcium influx kinetics in peripheral lymphocytes of patients with rheumatoid arthritis. Cytometry A. 83:287–293. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Kinsella S, Murphy K, Breen M, O'Neill S, McLaughlin P, Coyle J, Bogue C, O'Neill F, Moore N, McGarrigle A, et al: Comparison of single CT scan assessment of bone mineral density, vascular calcification and fat mass with standard clinical measurements in renal transplant subjects: The ABC HeART study. BMC Nephrol. 16:1882015. View Article : Google Scholar : PubMed/NCBI

28 

Morimoto Y, Hoshino H, Sakurai T, Terakawa S and Nagano A: Quantitative evaluation of bone resorption activity of osteoclast-like cells by measuring calcium phosphate resorbing area using incubator-facilitated and video-enhanced microscopy. Microsc Res Tech. 72:317–322. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Thomas MV and Puleo DA: Infection, inflammation, and bone regeneration: A paradoxical relationship. J Dent Res. 90:1052–1061. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Mallavia B, Recio C, Oguiza A, Ortiz-Muñoz G, Lazaro I, Lopez-Parra V, Lopez-Franco O, Schindler S, Depping R, Egido J and Gomez-Guerrero C: Peptide inhibitor of NF-κB translocation ameliorates experimental atherosclerosis. Am J Pathol. 182:1910–1921. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Xinluan W, Yuxiao L, Helena NH, Zhijun Y and Ling Q: Systemic drug delivery systems for bone tissue regeneration-a mini review. Curr Pharm Des. 21:1575–1583. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Gibbs DM, Black CR, Dawson JI and Oreffo RO: A review of hydrogel use in fracture healing and bone regeneration. J Tissue Eng Regen Med. 10:187–198. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Tomson PL, Lumley PJ, Alexander MY, Smith AJ and Cooper PR: Hepatocyte growth factor is sequestered in dentine matrix and promotes regeneration-associated events in dental pulp cells. Cytokine. 61:622–629. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Madonna R, Cevik C, Nasser M and De Caterina R: Hepatocyte growth factor: Molecular biomarker and player in cardioprotection and cardiovascular regeneration. Thromb Haemost. 107:656–661. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Jin SZ, Meng XW, Sun X, Han MZ, Liu BR, Wang XH and Pei FH: Hepatocyte growth factor promotes liver regeneration induced by transfusion of bone marrow mononuclear cells in a murine acute liver failure model. J Hepatobiliary Pancreat Sci. 18:397–405. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Izumida Y, Aoki T, Yasuda D, Koizumi T, Suganuma C, Saito K, Murai N, Shimizu Y, Hayashi K, Odaira M, et al: Hepatocyte growth factor is constitutively produced by donor-derived bone marrow cells and promotes regeneration of pancreatic beta-cells. Biochem Biophys Res Commun. 333:273–282. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Harvey EJ: Knee trauma and posttraumatic osteoarthritis-more science needed: Commentary on an article by Justin M. Haller, MD et al: ‘Inflammatory cytokine response following acute tibial plateau fracture’. J Bone Joint Surg Am. 97:e332015. View Article : Google Scholar : PubMed/NCBI

38 

Haller JM, McFadden M, Kubiak EN and Higgins TF: Inflammatory cytokine response following acute tibial plateau fracture. J Bone Joint Surg Am. 97:478–483. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Marino M, Palmieri G, Peruzzi M, Scuderi F and Bartoccioni E: A study of inflammatory/necrosis biomarkers in the fracture of the femur treated with proximal femoral nail antirotation. Mediators Inflamm. 2015:1898642015. View Article : Google Scholar : PubMed/NCBI

40 

Coudriet GM, He J, Trucco M, Mars WM and Piganelli JD: Hepatocyte growth factor modulates interleukin-6 production in bone marrow derived macrophages: Implications for inflammatory mediated diseases. PLoS One. 5:e153842010. View Article : Google Scholar : PubMed/NCBI

41 

Boissinot M, Cleyrat C, Vilaine M, Jacques Y, Corre I and Hermouet S: Anti-inflammatory cytokines hepatocyte growth factor and interleukin-11 are over-expressed in Polycythemia vera and contribute to the growth of clonal erythroblasts independently of JAK2V617F. Oncogene. 30:990–1001. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Katz MS, Thatch KA and Schwartz MZ: Hepatocyte growth factor and omega-3-enriched feeds have a synergistic effect on mucosal mass in an animal model of inflammatory bowel disease. J Pediatr Surg. 47:194–198. 2012. View Article : Google Scholar : PubMed/NCBI

43 

da Silva CG, Maccariello ER, Wilson SW, Putheti P, Daniel S, Damrauer SM, Peterson CR, Siracuse JJ, Kaczmarek E and Ferran C: Hepatocyte growth factor preferentially activates the anti-inflammatory arm of NF-κB signaling to induce A20 and protect renal proximal tubular epithelial cells from inflammation. J Cell Physiol. 227:1382–1390. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Sasikumar KP, Elavarasu S and Gadagi JS: The application of bone morphogenetic proteins to periodontal and peri-implant tissue regeneration: A literature review. J Pharm Bioallied Sci. 4 Suppl 2:S427–S430. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Bragdon B, Moseychuk O, Saldanha S, King D, Julian J and Nohe A: Bone morphogenetic proteins: A critical review. Cell Signal. 23:609–620. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Hustedt JW and Blizzard DJ: The controversy surrounding bone morphogenetic proteins in the spine: A review of current research. Yale J Biol Med. 87:549–561. 2014.PubMed/NCBI

47 

Dagostino PR, Whitmore RG, Smith GA, Maltenfort MG and Ratliff JK: Impact of bone morphogenetic proteins on frequency of revision surgery, use of autograft bone, and total hospital charges in surgery for lumbar degenerative disease: Review of the Nationwide inpatient sample from 2002 to 2008. Spine J. 14:20–30. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Imai Y, Terai H, Nomura-Furuwatari C, Mizuno S, Matsumoto K, Nakamura T and Takaoka K: Hepatocyte growth factor contributes to fracture repair by upregulating the expression of BMP receptors. J Bone Miner Res. 20:1723–1730. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Ye L, Lewis-Russell JM, Davies G, Sanders AJ, Kynaston H and Jiang WG: Hepatocyte growth factor up-regulates the expression of the bone morphogenetic protein (BMP) receptors, BMPR-IB and BMPR-II, in human prostate cancer cells. Int J Oncol. 30:521–529. 2007.PubMed/NCBI

50 

Okubo Y, Bessho K, Fujimura K, Kusumoto K, Ogawa Y and Iizuka T: Osteogenesis by recombinant human bone morphogenetic protein-2 at skeletal sites. Clin Orthop Relat Res. 1–301. 2000.

51 

Ozeç Y, Oztürk M, Kýlýç E, Yeler H, Göze F and Gümüş C: Effect of recombinant human bone morphogenetic protein-2 on mandibular distraction osteogenesis. J Craniofac Surg. 17:80–83. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Marinova-Mutafchieva L, Taylor P, Funa K, Maini RN and Zvaifler NJ: Mesenchymal cells expressing bone morphogenetic protein receptors are present in the rheumatoid arthritis joint. Arthritis Rheum. 43:2046–2055. 2000. View Article : Google Scholar : PubMed/NCBI

53 

Issa JP, do Nascimento C, Lamano T, Iyomasa MM, Sebald W and de Albuquerque RF Jr: Effect of recombinant human bone morphogenetic protein-2 on bone formation in the acute distraction osteogenesis of rat mandibles. Clin Oral Implants Res. 20:1286–1292. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Yonezawa H, Harada K, Ikebe T, Shinohara M and Enomoto S: Effect of recombinant human bone morphogenetic protein-2 (rhBMP-2) on bone consolidation on distraction osteogenesis: A preliminary study in rabbit mandibles. J Craniomaxillofac Surg. 34:270–276. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Wang J, Wang K, Shi Z and Zhang M: Osteoprotegerin mRNA/receptor activator of NF-kappaB ligand mRNA expressions in bone tissues of glucocorticoid-induced osteonecrosis of the femoral head. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 22:1161–1164. 2008.(In Chinese). PubMed/NCBI

56 

Sugimori K, Matsui K, Motomura H, Tokoro T, Wang J, Higa S, Kimura T and Kitajima I: BMP-2 prevents apoptosis of the N1511 chondrocytic cell line through PI3K/Akt-mediated NF-kappaB activation. J Bone Miner Metab. 23:411–419. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Feng JQ, Xing L, Zhang JH, Zhao M, Horn D, Chan J, Boyce BF, Harris SE, Mundy GR and Chen D: NF-kappaB specifically activates BMP-2 gene expression in growth plate chondrocytes in vivo and in a chondrocyte cell line in vitro. J Biol Chem. 278:29130–29135. 2003. 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
Zhen R, Yang J, Wang Y, Li Y, Chen B, Song Y, Ma G and Yang B: Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway. Mol Med Rep 17: 6045-6053, 2018
APA
Zhen, R., Yang, J., Wang, Y., Li, Y., Chen, B., Song, Y. ... Yang, B. (2018). Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway. Molecular Medicine Reports, 17, 6045-6053. https://doi.org/10.3892/mmr.2018.8559
MLA
Zhen, R., Yang, J., Wang, Y., Li, Y., Chen, B., Song, Y., Ma, G., Yang, B."Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway". Molecular Medicine Reports 17.4 (2018): 6045-6053.
Chicago
Zhen, R., Yang, J., Wang, Y., Li, Y., Chen, B., Song, Y., Ma, G., Yang, B."Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway". Molecular Medicine Reports 17, no. 4 (2018): 6045-6053. https://doi.org/10.3892/mmr.2018.8559