Open Access

Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis

  • Authors:
    • Cuisong Tang
    • Dengpan Liang
    • Yuyou Qiu
    • Jingqi Zhu
    • Guangyu Tang
  • View Affiliations

  • Published online on: February 17, 2022     https://doi.org/10.3892/mmr.2022.12648
  • Article Number: 132
  • Copyright: © Tang 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

Osteoporosis is a bone‑related disease that results from impaired bone formation and excessive bone resorption. The potential value of adipokines has been investigated previously, due to their influence on osteogenesis. However, the osteogenic effects induced by omentin‑1 remain unclear. The aim of the present study was to determine the regulatory effects of omentin‑1 on osteoblast viability and differentiation, as well as to explore the underlying molecular mechanism. The present study investigated the effects of omentin‑1 on the viability and differentiation of mouse pre‑osteoblast cells (MC3T3‑E1) using quantitative and qualitative measures. A Cell Counting Kit‑8 assay was used to assess the viability of MC3T3‑E1 cells following treatment with different doses of omentin‑1. Omentin‑1 and bone morphogenetic protein (BMP) inhibitor were added to osteogenic induction mediums in different ways to assess their effect. The alkaline phosphatase (ALP) activity and Alizarin Red S (ARS) staining of MC3T3‑E1 cells treated with omentin‑1 and/or BMP inhibitor were used to examine the effects of omentin‑1 on differentiation and mineralization. Western blotting was used to further explore its potential mechanism, and to study the role of omentin‑1 on the viability and differentiation of osteoblasts. The results showed that omentin‑1 altered the viability of MC3T3‑E1 cells in a dose‑dependent manner. Omentin‑1 treatment significantly increased the expression of members of the TGF‑β/Smad signaling pathway. In the omentin‑1 group, the ALP activity of the MC3T3‑E1 cells was increased, and the ARS staining area was also increased. The mRNA and protein expression levels of BMP2, Runt‑related transcription factor 2, collagen1, osteopontin, osteocalcin and osterix in the omentin‑1 group were also significantly upregulated. All these effects were reversed following treatment with SIS3 HCl. These results demonstrated that omentin‑1 can significantly promote osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway, thereby promoting bone formation and preventing osteoporosis.

Introduction

Osteoporosis is a debilitating disease that predominantly affects elderly individuals worldwide (1). At present, >200 million patients suffer from osteoporosis worldwide (2). According to research statistics, >1.6 million fractures are caused by osteoporosis (3,4). For women aged >50 years, the risk of suffering from fractures due to osteoporosis is 40–50% (5). Osteoporosis is a systemic skeletal disease that is characterized by a decrease in mass and deterioration of the bone microstructure (6). The consequences brought on by osteoporosis are bone fragility and susceptibility to fractures, and this can cause brittle fractures (7). At the cellular level, the pathogenesis of osteoporosis is primarily related to the disruption of bone remodeling (8). Bone remodeling primarily depends on osteoblast osteogenesis and osteoclast bone absorption, so as to maintain a certain balance (9). When a disease or aberrant inflammation disrupt this balance, osteoporosis may occur (10).

In bone tissues, osteoblasts originate from mesenchymal stem cells (MSCs), whereas osteoclasts are derived from hematopoietic stem cells (11). The differentiation of osteoblasts is achieved through the initial organic phase, which is production of collagen matrix, and the final inorganic phase, which is mineralization by hydroxyapatite crystals on the collagen scaffold (12). After the completion of matrix secretion and mineralization, the osteoblasts, which are embedded in the matrix, are known as bone cells, whereas the osteoblasts remaining on the bone surface turn into flattened lining cells or gradually undergo apoptosis. Importantly, bone marrow MSCs (BMSCs) are pluripotent. Apart from osteoblasts, BMSCs can differentiate into several different cell types, including adipocytes, myocytes, chondrocytes, endothelial cells and vascular smooth muscle cells (13). Amongst these cell lineages, adipogenic differentiation is particularly relevant to the homeostasis of bone, as cells undergoing osteoblast formation can be diverted to adipocytes, leading to the loss of these cells from the osteoblast pool and the reduction of overall osteogenic potential (13).

The imbalance of adipocyte differentiation and regulation of osteoblasts is commonly seen in aging and diabetic individuals, which can lead to the impairment of fat bone marrow, impairment of osteoblast renewal and an increase in the incidence of chronic bone loss (1315). Beyond that, adipocytes secrete a series of biologically active signaling molecules, which can also influence bone homeostasis. It has been reported that several of these molecules, including chemorin, resistin, visfatin, leptin and adiponectin, affect the development and function of osteoblasts and osteoclasts (1621).

Omentin-1, a 34 kDa adipokine selectively expressed in omental adipose tissue, is abundant in the plasma (22,23). Omentin-1 has been demonstrated to serve as a significant factor in multiple physiological processes, including insulin action, cardiovascular function and the inflammatory response (2325). A previous study demonstrated that omentin-1 plays an important role in protecting against vascular calcification (26). A clinical study has also shown that omentin-1 levels are inversely correlated with obesity and insulin resistance (27). Regarding its effects on bone, a previous study reported that circulating omentin-1 levels were inversely correlated with bone mineral density (BMD) in the lumbar region of the spine in Iranian postmenopausal women (28). The levels of circulating omentin-1 correlates positively with adiponectin and negatively with body mass index and the leptin levels (29). In a co-culture system of osteoblasts and osteoclast precursors, omentin-1 can reduce osteoclast formation by stimulating osteoprotegerin (OPG) (30). To date, only a few studies have investigated the association between omentin-1 and osteoblast proliferation and differentiation. Moreover, the osteogenic effect induced by omentin-1 remains unclear. The TGF-β/Smad signaling pathway is involved in the regulation of cell differentiation and it is a key pathway related to osteogenesis (3133). To understand the underlying mechanism driving the effects of omentin-1 on osteogenic differentiation in MC3T3-E1, the role of the TGF-β/Smad pathway in this process was investigated. The aim of the present study was to determine the regulatory effects of omentin-1 on osteoblast viability and differentiation, as well as to explore the underlying molecular mechanism.

Materials and methods

Cell culture and treatment

Mouse embryo osteoblast precursor cell line MC3T3-E1 was obtained from American Type Culture Collection (ATCC, CRL-2593™). The cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (HyClone; Cytiva) and 1% penicillin/streptomycin (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 humidified incubator. In order to stimulate osteogenic induction, the MC3T3-E1 cells were seeded in DMEM supplemented with 10% FBS, 1% penicillin/streptomycin, 10 mM β-glycerophosphate, 50 µg/ml ascorbic acid and 10 nM dexamethasone (all Sigma-Aldrich; Merck KGaA) (34). Subsequently, cells were treated with different doses of omentin-1 (100, 500 and 1,000 ng/ml; Cell Science, Inc.) for 24 h at 37°C and/or 3 µM SIS3 HCl (Selleck Chemicals) for 6 h at 37°C for the following experiments.

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted from cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). iScript™ Reverse Transcription SuperMix kit (Bio-Rad Laboratories, Inc.) was used to reverse transcribe 2 µg RNA into cDNA according to the manufacturer's protocol. qPCR was performed using SYBR-Green MasterMix (Applied Biosystems; Thermo Fisher Scientific, Inc.) on an ABI PRISM 7900 Sequence Detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The PCR amplification program was as follows: 94°C for 60 sec, followed by 40 cycles of 94°C for 30 sec, 60°C for 30 sec and 72°C for 60 sec. The primer sequences used for PCR were as follows: Bone morphogenetic protein 2 (BMP2) forward, 5′-AGCCCTTCACTGCCATCCTGT-3′ and reverse, 5′-ATTCTCTCGTTCACCGCCCAC-3′; Runt-related transcription factor 2 (Runx2) forward, 5′-CCCAACTTCCTGTGCTCC-3′ and reverse, 5′-AGTGAAACTCTTGCCTCGTC-3′; Collagen1 forward, 5′-TAAGGGTGACAGAGGCGATG-3′ and reverse, 5′-GGACCGCTAGGACCAGTTTC-3′; osteopontin (Opn) forward, 5′-TCCAAAGTCAGCCAGGAATCC-3′ and reverse, 5′-CGGAGTTGTCTGTGCTCTTCA-3′; osteocalcin (Ocn) forward, 5′-CTCCTTACCCGGATCCCCTG-3′ and reverse, 5′-GTAGAAGCGCTGGTAGGCGT-3′; Osterix forward, 5′-TCCCTGGATATGACTCATCCCT-3′ and reverse, 5′-CCAAGGAGTAGGTGTGTTGCC-3′; and GAPDH forward, 5′-GGGAAACTGTGGCGTGAT-3′ and reverse, 5′-GAGTGGGTGTCGCTGTTGA-3′. Gene expression analysis was performed using the 2−ΔΔCq method (35) and normalized to GAPDH.

Cell viability

The cells were cultured in 96-well culture plates at a density of 2×104 cells/well. After 3 days, a Cell Counting Kit-8 (CCK-8) assay (Beyotime Institute of Biotechnology) was performed to evaluate cell viability. The CCK-8 solution was added to each group, and the cells were incubated at 37°C for 1 h. The absorbance of each well was measured at a wavelength of 450 nm using a microplate spectrophotometer.

Western blot assay

Cells in the different treatment groups were lysed using RIPA lysis buffer (Beyotime Institute of Biotechnology) and protein concentrations were measured using Bradford reagent (Bio-Rad Laboratories, Inc.). Equal quantities of proteins (40 µg/lane) were separated via 10% SDS-PAGE (Bio-Rad Laboratories, Inc.) and transferred to a PVDF membrane (MilliporeSigma). After being blocked with 5% non-fat milk in 0.1% tris-buffered saline with Tween-20 for 1 h at room temperature, the membranes were incubated with primary antibodies against BMP2 (1:1,000; cat. no. ab214821; Abcam), phosphorylated (p)-Smad1 (1:1,000; cat. no. ab226821; Abcam), p-Smad5 (1:2,000; cat. no. ab92698; Abcam), Smad1 (1:1,000; cat. no. ab33902; Abcam), Smad5 (1:1,000; cat. no. ab40771; Abcam), Runx2 (1:1,000; cat. no. ab236639; Abcam), collagen1 (1:1,000; cat. no. ab138492; Abcam), Opn (1:1,000; cat. no. ab63856; Abcam), Ocn (1:1,000; cat. no. ab133612; Abcam) and osterix (1:1,000; cat. no. ab209484; Abcam) and GAPDH (1:2,500; cat. no. ab9485; Abcam) overnight at 4°C, and then incubated with the appropriate horseradish peroxidase-conjugated secondary antibody (1:1,000; cat. no. #7074; Cell Signaling Technology, Inc.) for 1 h at room temperature. The blots were then visualized using an enhanced chemiluminescence system (Beyotime Institute of Biotechnology). Densitometry analysis was performed using ImageJ (Version 1.49; National Institutes of Health).

Cell differentiation analysis

Alkaline phosphatase (ALP) activity, an early biochemical marker widely used to assess osteogenic activity, was measured. Briefly, the cells were seeded in 12-well culture plates at a density of 4×104 cells per ml for 7 days. Subsequently, the cells were fixed in 4% paraformaldehyde for 30 min at room temperature, followed by treatment with nitroblue tetrazolium (Sigma-Aldrich; Merck KGaA) and 5-bromo-4-chloro-3-indolyl phosphate (Sigma-Aldrich; Merck KGaA) for 2 h at room temperature. Cells were washed with deionized water and observed under an inverted light microscope (Nikon Corporation; ×200 magnification).

Cell mineralization analysis

Mineralization was evaluated by quantifying the formation of calcium phosphate in cells using an Alizarin Red Staining (ARS) kit (Sigma-Aldrich; Merck KGaA). Briefly, the cells were cultured in 12-well culture plates at a density of 4×104 cells per ml. After 21 days, the cultured cells were fixed with 95% ethanol for 10 min at room temperature. To stain the calcium deposits, 2% ARS solution (Sigma-Aldrich; Merck KGaA) was applied for 15 min at room temperature. To measure the degree of mineralization, the ARS released from the cell matrix was incubated in cetyl pyridinium chloride for 15 min at room temperature and quantified by spectrophotometry at 540 nm.

Statistical analysis

Data are presented as the mean ± standard deviation of three independent experiments. Differences between groups were determined using an unpaired two-tailed Student's t-test for comparisons between two groups, and one-way ANOVA with a post hoc Tukey's test was used for comparisons between multiple groups. Statistical analysis was performed using SPSS 13.0 (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Omentin-1 stimulates viability of osteoblasts

The effect of omentin-1 on MC3T3-E1 cell viability was assessed using a CCK-8 assay. As shown in Fig. 1, the viability of osteoblasts was altered in a dose-dependent manner following treatment with omentin-1, and the effect was significant when treated with 1,000 ng/ml omentin-1 after 3 days of treatment.

Omentin-1 activates the Smad signaling pathway and increases BMP2 expression in osteoblasts

To explore the potential mechanisms by which omentin-1 exerted its effects on osteoblasts, western blotting was performed. As shown in Fig. 2, omentin-1 at a dose of 1,000 ng/ml increased the protein expression levels of BMP2, p-Smad1 and p-Smad5 (P<0.05). However, 1,000 ng/ml omentin-1 did not exert a notable effect on the expression levels of Smad1 and Smad5.

Omentin-1 promotes the viability of osteoblasts through the TGF-β/Smad signaling pathway

As shown in Fig. 3A and B, SIS3 HCl, a Smad inhibitor, significantly inhibited BMP2 protein levels and phosphorylation of Smad1 and Smad5 compared with the control group, while 1,000 ng/ml omentin-1 significantly increased the expression levels of the three proteins. Meanwhile, SIS3 HCl attenuated the effects of omentin-1 on the expression of BMP2, p-Smad1 and p-Smad5 in MC3T3-E1 cells. In addition, CCK-8 analysis showed a significant increase in cell viability in omentin-1-treated MC3T3-E1 cells, and this increase in viability was significantly attenuated by SIS3 HCl (Fig. 3C).

Omentin-1 regulates differentiation and mineralization of osteoblasts via the TGFβ/Smad signaling pathway

Next, the effects of omentin-1 on osteoblasts were assessed. As shown in Fig. 4A and B, ARS staining in the omentin-1-treated cells showed an increased mineralization rate compared with the control group, and SIS3 HCl significantly attenuated the omentin-1-induced mineralization. In addition, ALP activity was significantly increased in the omentin-1 group, whereas SIS3 HCl significantly weakened the activity of ALP in MC3T3-E1 cells (Fig. 4C).

Analysis of cellular Runx2, collagen1, Opn, Ocn and osterix expression

Data from RT-qPCR and western blot assays showed that the expression levels of osteogenesis-related proteins, including BMP2, Runx2, collagen1, Opn, Ocn and osterix, were significantly upregulated in the omentin-1 group. However, the expression levels of these proteins in MC3T3-E1 cells treated with SIS3 HCl were significantly reduced (Fig. 5A-C).

Discussion

Bone remodeling and skeletal homeostasis are dynamic life-long processes that rely on the balance and integrated actions between osteoblastic bone formation and osteoclastic bone resorption (3638). Osteoblastic lineage cells interact with osteoclastic lineage cells, which is necessary for the formation of functional osteoclasts (39). Certain bone-related diseases, such as osteoporosis, rheumatoid arthritis and osteoarthritis, are associated with abnormal bone metabolism and accelerated bone loss (40). Bone formation and repair are intrinsically associated with the balanced activity of osteoblasts (41). Osteoblasts are responsible for bone matrix secretion and mineralization, and they tightly regulate osteoclast activation and differentiation (42). Dysfunctional behaviors of osteoblasts lead to improper bone matrix deposition and mineralization, affecting the size, shape and integrity of the skeletal structures (43).

MC3T3-E1, a popular osteoblast cell line that represents a pre-osteoblastic phenotype, is a mouse calvaria clonal cell line that has given rise to several subclones (44). Decreased proliferative activities have been observed at passage numbers above 36, whereas their replicative senescence has been displayed when these cells reach a high passage number of 60, which is similar to that of human cells, at which point they show inconsistent cell cycling (45). This important feature also makes these cells suitable as novel models for in vitro bone research, including bone remodeling and formation (46).

Omentin-1, which was discovered from a human omental fat cDNA library, is primarily expressed in visceral adipose tissue (23). Omentin-1 can promote osteogenic differentiation, although this has been contested (4752). Therefore, the present study investigated the effects of omentin-1 on MC3T3-EI cells by observing proliferation patterns, with the aim of determining the regulatory effect of omentin-1 and identifying the underlying molecular mechanisms. When MC3T3-E1 cells were treated with different concentrations of omentin-1 (100-1,000 ng/ml) for 3 days, the viability of MC3T3-E1 cells increased significantly. Additionally, the effect of omentin-1 was weakened after adding a BMP inhibitor. This suggested that omentin-1 could stimulate the viability of osteoblasts. Based on these experiments, 1,000 ng/ml omentin-1 was used for subsequent experiments, and 0 ng/ml omentin-1 was used as the control group to further study the effects of omentin-1 on osteoblasts.

Regulation of gene expression by TGF-β is achieved primarily through the initiation of intracellular signal transduction and the activation of the Smad signaling pathway (53,54). TGF-β promotes osteogenic differentiation, and at the same time, it also inhibits adipogenic differentiation of human MSCs (55). The TGF-β/Smad pathway is also key to adipogenesis (56). TGF-β inhibits adipogenic differentiation in human preadipocytes, which is mediated primarily by the Smad family members (57,58). Smad3 inhibits adipogenic conversion, whereas interfering with Smad3 function confers resistance to inhibition of adipogenesis by TGF-β (56). The present results showed that SIS3 HCL treatment significantly inhibited the protein expression levels of BMP2, p-SMAD1 and p-SMAD5, while 1,000 ng/ml omentin-1 increased the protein expression levels of BMP2 and p-Smad1/5. However, 1,000 ng/ml omentin-1 exerted no effect on Smad1 and Smad5, which are also members of the TGF-β/Smad pathway.

To confirm the effects of the omentin-1/TGF-β/Smad pathway on MC3T3-E1 cells, cells were pretreated with a selective BMP TGF-β/Smad signaling inhibitor, SIS3 HCl (3 µM per day), 3 days prior to treatment with 1,000 ng/ml omentin-1. Cell viability was promoted in the 1,000 ng/ml omentin-1 group, and SIS3 HCl attenuated the increase in viability mediated by omentin-1. The results from the CCK-8 assays revealed that omentin-1 significantly promoted the viability ability of the MC3T3-E1 cells via the TGF-β/Smad pathway.

In previous years, several studies have investigated the relationship between omentin-1 and bone metabolism. On the one hand, adiponectin has been shown to induce osteoblast proliferation and differentiation (50), as well as to increase bone mass via osteoclastogenesis suppression and osteoblastogenesis activation (51), and the results of the present study were in agreement with these previous findings. Conversely, adiponectin has been verified to induce osteoclast formation via stimulation of RANKL and inhibition of OPG production (48,49). This suggests that adipokines also serve an important role in osteoclastic balance; thus the effects of adipokines on osteoclasts should form the basis of future studies. A previous study showed that there was an inverse correlation between omentin-1 and broadband ultrasound attenuation levels in postmenopausal women. This association was not mediated by OPG levels (52). The results from in vitro and in vivo studies have shown contradictory results. Results from a meta-analysis of observational studies demonstrated an inverse relationship between adiponectin levels and BMD, with five studies showing a negative association, and another five showing no association (47). The reasons for such discrepant findings are unclear, but it is conceivable that the microenvironment in human bodies can be affected by various factors. The ability of cell culture and animal models to emulate the complex processes that take place in the human body is limited, such as possible adiponectin-dependent counter-regulatory mechanisms.

Osteoporosis, a type of bone-related disease, results from impaired bone formation and excessive bone resorption. Current strategies to augment bone formation, such as the use of growth factors (59), have shown promising results. However, the osteoblast lineage specification and bone regeneration remain unclear. An improved understanding of these may considerably improve the therapeutic strategies available. In order to determine how omentin-1 guides osteoblast bone formation, the effects of several potential mediators involved in the pro-osteogenic pathway were assessed using western blotting in the current study. Osteoblasts are derived from osteoprogenitors that differentiate by progressively expressing maturation markers (60). Runx2 is a transcription factor that serves a role in osteoblast differentiation. Previous studies have shown that silencing of Runx2 can block the differentiation of osteoblasts in mice (61,62). Patients with cleidocranial dysplasia, which is caused by heterozygous mutations in the Runx2 gene, are characterized by an underdeveloped collarbone, short stature, excess teeth, fontanelle patency and other bone growth-related defects (63). Osterix is a zinc finger transcription factor present in osteoblasts and serves a leading role in the osteoblast differentiation process (63). A previous study showed that Runx2 can regulate the levels of osterix (64). Collagen-1 is a primary structural protein present in the extracellular space in the bone, making up 25–35% of the entire-body protein content (65). Depending upon the degree of mineralization, collagen tissues may be rigid (bone), compliant (tendon) or have a gradient from rigid to compliant (cartilage) (66). As the skeleton is the main structural component of the body, collagen-1 is responsible for maintaining the strength of these structures (67). Opn is also known as bone sialoprotein I, a protein in humans encoded by the SPP1 gene (68). Ocn is a vitamin K-dependent calcium-binding protein (69). Opn and Ocn are two typical biomarkers of osteoblasts, and are involved in osteogenic differentiation and the mineralization of extracellular matrix during bone formation and repair (70,71). Previous studies have shown that Runx2 stimulates the differentiation of MSCs into osteoblasts by regulating Opn and Ocn activity (61,72). In the present study, it was shown that omentin-1 significantly upregulated the expression of Runx2, Collagen-1, Opn, Ocn and osterix, suggesting that it accelerated bone formation.

Based on the aforementioned results, it was noticed in the present study that SIS3 HCL treatment alone only affected the TGF-β/Smad pathway, but had no significant effects on the viability, differentiation and mineralization of osteoblasts and the expression of osteogenesis-related proteins. However, SIS3 HCL exerted obvious roles when the cells were treated with omentin-1. Hence, we speculated that under normal conditions, osteoblasts were only partially affected by SIS3 HCL except for the expression of TGF-β/Smad pathway, but omentin-1 had significant promoting effects on cell differentiation and mineralization via regulating the TGF-β/Smad pathway. Thus, there is a noticeable effect of SIS3 HCL on biological function and signaling and omentin-1-mediated osteoblasts.

In conclusion, the present study examined the potential effects of omentin-1 on the viability and differentiation of osteoblasts and the signaling pathways involved (Fig. 6). Omentin-1 promoted osteoblast viability in a dose-dependent manner. Western blotting revealed that omentin-1 induced the activation of BMP2 and p-Smad1/5. Furthermore, omentin-1 promoted osteoblast viability, differentiation and mineralization, and these effects were impeded by a TGF-β/Smad inhibitor. These findings indicated that omentin-1 promoted osteoblast viability and differentiation via the TGF-β/Smad signaling pathway, suggesting that omentin-1 may be a potential target in the treatment of osteoporosis.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

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

Authors' contributions

CT, DL and GT designed the study, drafted and revised the manuscript. YQ and JZ analyzed the data and searched the literature. CT, DL, YQ and JZ performed the experiments. CT and GT confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Miller PD: Management of severe osteoporosis. Expert Opin Pharmacother. 17:473–488. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Noh JY, Yang Y and Jung H: Molecular mechanisms and emerging therapeutics for osteoporosis. Int J Mol Sci. 21:76232020. View Article : Google Scholar : PubMed/NCBI

3 

Clark S: Osteoporosis-the disease of the 21st century? Lancet. 359:17142002. View Article : Google Scholar : PubMed/NCBI

4 

Cooper C: Epidemiology of osteoporosis. Osteoporos Int. 9 (Suppl 2):S2–S8. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Johnell O and Kanis J: Epidemiology of osteoporotic fractures. Osteoporos Int. 16 (Suppl 2):S3–S7. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Straka M, Straka-Trapezanlidis M, Deglovic J and Varga I: Periodontitis and osteoporosis. Neuro Endocrinol Lett. 36:401–406. 2015.PubMed/NCBI

7 

Kurra S, Fink DA and Siris ES: Osteoporosis-associated fracture and diabetes. Endocrinol Metab Clin North Am. 43:233–243. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Lane NE: Epidemiology, etiology, and diagnosis of osteoporosis. Am J Obstet Gynecol. 194 (Suppl 2):S3–S11. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Baccaro LF, Conde DM, Costa-Paiva L and Pinto-Neto AM: The epidemiology and management of postmenopausal osteoporosis: A viewpoint from Brazil. Clin Interv Aging. 10:583–591. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Lane JM, Russell L and Khan SN: Osteoporosis. Clin Orthop Relat Res. 372:139–150. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Chen X, Wang Z, Duan N, Zhu G, Schwarz EM and Xie C: Osteoblast-osteoclast interactions. Connect Tissue Res. 59:99–107. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Jayakumar P and Di Silvio L: Osteoblasts in bone tissue engineering. Proc Inst Mech Eng H. 224:1415–1440. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Malhotra A and Habibovic P: Calcium phosphates and angiogenesis: Implications and advances for bone regeneration. Trends Biotechnol. 34:983–992. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Leijten J, Chai YC, Papantoniou I, Geris L, Schrooten J and Luyten FP: Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev. 84:30–44. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wei DX, Dao JW and Chen GQ: A Micro-Ark for Cells: Highly open porous polyhydroxyalkanoate microspheres as injectable scaffolds for tissue regeneration. Adv Mater. 30:e18022732018. View Article : Google Scholar : PubMed/NCBI

16 

Shen X, Zhang Y, Gu Y, Xu Y, Liu Y, Li B and Chen L: Sequential and sustained release of SDF-1 and BMP-2 from silk fibroin-nanohydroxyapatite scaffold for the enhancement of bone regeneration. Biomaterials. 106:205–216. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Cahill KS, Chi JH, Day A and Claus EB: Prevalence, complications, and hospital charges associated with use of bone-morphogenetic proteins in spinal fusion procedures. JAMA. 302:58–66. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Skovrlj B, Koehler SM, Anderson PA, Qureshi SA, Hecht AC, Iatridis JC and Cho SK: Association between BMP-2 and carcinogenicity. Spine (Phila Pa 1976). 40:1862–1871. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Xu C, Su P, Chen X, Meng Y, Yu W, Xiang AP and Wang Y: Biocompatibility and osteogenesis of biomimetic Bioglass-Collagen-Phosphatidylserine composite scaffolds for bone tissue engineering. Biomaterials. 32:1051–1058. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Zhang Y, Fan W, Ma Z, Wu C, Fang W, Liu G and Xiao Y: The effects of pore architecture in silk fibroin scaffolds on the growth and differentiation of mesenchymal stem cells expressing BMP7. Acta Biomater. 6:3021–3028. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Holzwarth JM and Ma PX: Biomimetic nanofibrous scaffolds for bone tissue engineering. Biomaterials. 32:9622–9629. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Schaffler A, Neumeier M, Herfarth H, Furst A, Scholmerich J and Buchler C: Genomic structure of human omentin, a new adipocytokine expressed in omental adipose tissue. Biochim Biophys Acta. 1732:96–102. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC and Gong DW: Identification of omentin as a novel depot-specific adipokine in human adipose tissue: Possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 290:E1253–E1261. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Yamawaki H, Kuramoto J, Kameshima S, Usui T, Okada M and Hara Y: Omentin, a novel adipocytokine inhibits TNF-induced vascular inflammation in human endothelial cells. Biochem Biophys Res Commun. 408:339–343. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Yamawaki H, Tsubaki N, Mukohda M, Okada M and Hara Y: Omentin, a novel adipokine, induces vasodilation in rat isolated blood vessels. Biochem Biophys Res Commun. 393:668–672. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Duan XY, Xie PL, Ma YL and Tang SY: Omentin inhibits osteoblastic differentiation of calcifying vascular smooth muscle cells through the PI3K/Akt pathway. Amino Acids. 41:1223–1231. 2011. View Article : Google Scholar : PubMed/NCBI

27 

de Souza Batista CM, Yang RZ, Lee MJ, Glynn NM, Yu DZ, Pray J, Ndubuizu K, Patil S, Schwartz A, Kligman M, et al: Omentin plasma levels and gene expression are decreased in obesity. Diabetes. 56:1655–1661. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Tohidi M, Akbarzadeh S, Larijani B, Kalantarhormozi M, Ostovar A, Assadi M, Vahdat K, Farrokhnia M, Sanjdideh Z, Amirinejad R and Nabipour I: Omentin-1, visfatin and adiponectin levels in relation to bone mineral density in Iranian postmenopausal women. Bone. 51:876–881. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Moreno-Navarrete JM, Catalán V, Ortega F, Gómez-Ambrosi J, Ricart W, Frühbeck G and Fernández-Real JM: Circulating omentin concentration increases after weight loss. Nutr Metab (Lond). 7:272010. View Article : Google Scholar : PubMed/NCBI

30 

Xie H, Xie PL, Wu XP, Chen SM, Zhou HD, Yuan LQ, Sheng ZF, Tang SY, Luo XH and Liao EY: Omentin-1 attenuates arterial calcification and bone loss in osteoprotegerin-deficient mice by inhibition of RANKL expression. Cardiovasc Res. 92:296–306. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Wang J, Wang M, Chen F, Wei Y, Chen X, Zhou Y, Yang X, Zhu X, Tu C and Zhang X: Nano-hydroxyapatite coating promotes porous calcium phosphate ceramic-induced osteogenesis via BMP/Smad signaling pathway. Int J Nanomedicine. 14:7987–8000. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Liu Z, Yu Z, Chang H, Wang Y, Xiang H, Zhang X and Yu B: Strontium-containing α-calcium sulfate hemihydrate promotes bone repair via the TGF-β/Smad signaling pathway. Mol Med Rep. 20:3555–3564. 2019.PubMed/NCBI

33 

Wang QL, Li HF, Wang DP, Liu ZY, Xiao WW, Xu LL and Yu S: Effect of GGCX on the differentiation function of osteoporosis bone marrow mesenchymal stem cells through regulating TGFβ/smad signaling pathway. Eur Rev Med Pharmacol Sci. 23:7224–7231. 2019.PubMed/NCBI

34 

Kim SW, Her SJ, Park SJ, Kim D, Park KS, Lee HK, Han BH, Kim MS, Shin CS and Kim SY: Ghrelin stimulates proliferation and differentiation and inhibits apoptosis in osteoblastic MC3T3-E1 cells. Bone. 37:359–369. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Lerner UH, Kindstedt E and Lundberg P: The critical interplay between bone resorbing and bone forming cells. J Clin Periodontol. 46 (Suppl 21):33–51. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Fili S, Karalaki M and Schaller B: Mechanism of bone metastasis: The role of osteoprotegerin and of the host-tissue microenvironment-related survival factors. Cancer Lett. 283:10–19. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Kearns AE, Khosla S and Kostenuik PJ: Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulation of bone remodeling in health and disease. Endocr Rev. 29:155–192. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Horowitz MC, Xi Y, Wilson K and Kacena MA: Control of osteoclastogenesis and bone resorption by members of the TNF family of receptors and ligands. Cytokine Growth Factor Rev. 12:9–18. 2001. View Article : Google Scholar : PubMed/NCBI

40 

Wada T, Nakashima T, Hiroshi N and Penninger JM: RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 12:17–25. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Long F: Building strong bones: Molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol. 13:27–38. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Nakamichi Y, Udagawa N, Kobayashi Y, Nakamura M, Yamamoto Y, Yamashita T, Mizoguchi T, Sato M, Mogi M, Penninger JM and Takahashi N: Osteoprotegerin reduces the serum level of receptor activator of NF-kappaB ligand derived from osteoblasts. J Immunol. 178:192–200. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Valenti MT, Dalle Carbonare L and Mottes M: Osteogenic differentiation in healthy and pathological conditions. Int J Mol Sci. 18:412016. View Article : Google Scholar : PubMed/NCBI

44 

Wan Hasan WN, Abd Ghafar N, Chin KY and Ima-Nirwana S: Annatto-derived tocotrienol stimulates osteogenic activity in preosteoblastic MC3T3-E1 cells: A temporal sequential study. Drug Des Devel Ther. 12:1715–1726. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Czekanska EM, Stoddart MJ, Richards RG and Hayes JS: In search of an osteoblast cell model for in vitro research. Eur Cell Mater. 24:1–17. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Aubin JE: Regulation of osteoblast formation and function. Rev Endocr Metab Disord. 2:81–94. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Biver E, Salliot C, Combescure C, Gossec L, Hardouin P, Legroux-Gerot I and Cortet B: Influence of Adipokines and Ghrelin on Bone Mineral Density and Fracture Risk: A systematic review and meta-analysis. J Clin Endocrinol Metab. 96:2703–2713. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Luo XH, Guo LJ, Xie H, Yuan LQ, Wu XP, Zhou HD and Liao EY: Adiponectin stimulates RANKL and inhibits OPG expression in human osteoblasts through the MAPK signaling pathway. J Bone Miner Res. 21:1648–1656. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Wang QP, Li XP, Wang M, Zhao LL, Li H, Xie H and Lu ZY: Adiponectin exerts its negative effect on bone metabolism via OPG/RANKL pathway: An in vivo study. Endocrine. 47:845–853. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Luo XH, Guo LJ, Yuan LQ, Xie H, Zhou HD, Wu XP and Liao EY: Adiponectin stimulates human osteoblasts proliferation and differentiation via the MAPK signaling pathway. Exp Cell Res. 309:99–109. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Oshima K, Nampei A, Matsuda M, Iwaki M, Fukuhara A, Hashimoto J, Yoshikawa H and Shimomura I: Adiponectin increases bone mass by suppressing osteoclast and activating osteoblast. Biochem Biophys Res Commun. 331:520–526. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Wu SS, Liang QH, Liu Y, Cui RR, Yuan LQ and Liao EY: Omentin-1 stimulates human osteoblast proliferation through PI3K/Akt signal pathway. Int J Endocrinol. 2013:3689702013. View Article : Google Scholar : PubMed/NCBI

53 

Miyazono K: TGF-beta signaling by Smad proteins. Cytokine Growth Factor Rev. 11:15–22. 2000. View Article : Google Scholar : PubMed/NCBI

54 

ten Dijke P, Miyazono K and Heldin CH: Signaling inputs converge on nuclear effectors in TGF-beta signaling. Trends Biochem Sci. 25:64–70. 2000. View Article : Google Scholar : PubMed/NCBI

55 

van Zoelen EJ, Duarte I, Hendriks JM and van der Woning SP: TGFβ-induced switch from adipogenic to osteogenic differentiation of human mesenchymal stem cells: Identification of drug targets for prevention of fat cell differentiation. Stem Cell Res Ther. 7:1232016. View Article : Google Scholar : PubMed/NCBI

56 

Santibanez JF and Kocic J: Transforming growth factor-β superfamily, implications in development and differentiation of stem cells. Biomol Concepts. 3:429–445. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Petruschke T, Röhrig K and Hauner H: Transforming growth factor beta (TGF-beta) inhibits the differentiation of human adipocyte precursor cells in primary culture. Int J Obes Relat Metab Disord. 18:532–536. 1994.PubMed/NCBI

58 

Choy L, Skillington J and Derynck R: Roles of autocrine TGF-beta receptor and Smad signaling in adipocyte differentiation. J Cell Biol. 149:667–682. 2000. View Article : Google Scholar : PubMed/NCBI

59 

El Bialy I, Jiskoot W and Reza Nejadnik M: Formulation, delivery and stability of bone morphogenetic proteins for effective bone regeneration. Pharm Res. 34:1152–1170. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Rutkovskiy A, Stensløkken KO and Vaage IJ: Osteoblast differentiation at a glance. Med Sci Monit Basic Res. 22:95–106. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Yang D, Okamura H and Qiu L: Upregulated osterix expression elicited by Runx2 and Dlx5 is required for the accelerated osteoblast differentiation in PP2A Cα-knockdown cells. Cell Biol Int. 42:403–410. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Komori T: Runx2, an inducer of osteoblast and chondrocyte differentiation. Histochem Cell Biol. 149:313–323. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Mundlos S, Otto F, Mundlos C, Mulliken JB, Aylsworth AS, Albright S, Lindhout D, Cole WG, Henn W, Knoll JH, et al: Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia. Cell. 89:773–779. 1997. View Article : Google Scholar : PubMed/NCBI

64 

Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR and de Crombrugghe B: The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 108:17–29. 2002. View Article : Google Scholar : PubMed/NCBI

65 

Varma S, Orgel JP and Schieber JD: Nanomechanics of type I collagen. Biophys J. 111:50–56. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Murshed M: Mechanism of bone mineralization. Cold Spring Harb Perspect Med. 8:a0312292018. View Article : Google Scholar : PubMed/NCBI

67 

Kisling A, Lust RM and Katwa LC: What is the role of peptide fragments of collagen I and IV in health and disease? Life Sci. 228:30–34. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Chen Y, Ou Y, Dong J, Yang G, Zeng Z, Liu Y, Liu B, Li W, He X and Lan T: Osteopontin promotes collagen I synthesis in hepatic stellate cells by miRNA-129-5p inhibition. Exp Cell Res. 362:343–348. 2018. View Article : Google Scholar : PubMed/NCBI

69 

An J, Yang H, Zhang Q, Liu C, Zhao J, Zhang L and Chen B: Natural products for treatment of osteoporosis: The effects and mechanisms on promoting osteoblast-mediated bone formation. Life Sci. 147:46–58. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Zhou H, Choong P, McCarthy R, Chou ST, Martin TJ and Ng KW: In situ hybridization to show sequential expression of osteoblast gene markers during bone formation in vivo. J Bone Miner Res. 9:1489–1499. 1994. View Article : Google Scholar : PubMed/NCBI

71 

van Leeuwen JP, van Driel M, van den Bemd GJ and Pols HA: Vitamin D control of osteoblast function and bone extracellular matrix mineralization. Crit Rev Eukaryot Gene Expr. 11:199–226. 2001.PubMed/NCBI

72 

Takahashi T, Kato S, Suzuki N, Kawabata N and Takagi M: Autoregulatory mechanism of Runx2 through the expression of transcription factors and bone matrix proteins in multipotential mesenchymal cell line, ROB-C26. J Oral Sci. 47:199–207. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2022
Volume 25 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
Tang C, Liang D, Qiu Y, Zhu J and Tang G: Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis. Mol Med Rep 25: 132, 2022
APA
Tang, C., Liang, D., Qiu, Y., Zhu, J., & Tang, G. (2022). Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis. Molecular Medicine Reports, 25, 132. https://doi.org/10.3892/mmr.2022.12648
MLA
Tang, C., Liang, D., Qiu, Y., Zhu, J., Tang, G."Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis". Molecular Medicine Reports 25.4 (2022): 132.
Chicago
Tang, C., Liang, D., Qiu, Y., Zhu, J., Tang, G."Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis". Molecular Medicine Reports 25, no. 4 (2022): 132. https://doi.org/10.3892/mmr.2022.12648