Mangiferin promotes the osteogenic differentiation of human periodontal ligament stem cells via TGF‑β/SMAD2 signaling

  • Authors:
    • Yingzhi Gu
    • Li Zhang
    • Yuxing Bai
  • View Affiliations

  • Published online on: June 24, 2022     https://doi.org/10.3892/mmr.2022.12782
  • Article Number: 266
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mangiferin (MAG) is a polyphenolic compound present in mangoes. This compound suppresses inflammation and decreases bone destruction. This study aimed to determine whether MAG directly promotes proliferation and osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs). Cell proliferation and osteogenic differentiation experiments were performed in hPDLSCs, and MAG was used as a stimulator during osteogenic induction. Alkaline phosphatase (ALP) activity and Alizarin red staining were analyzed, and the expression of osteogenesis‑associated genes was investigated by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analysis to determine the effect of MAG on the osteogenic differentiation of hPDLSCs. Galunisertib was used to selectively inhibit TGF‑β/SMAD2 signaling. Western blotting was performed to study the underlying mechanism. Cell Counting Kit‑8 assay showed that MAG did not promote the proliferation of hPDLSCs. MAG (200 µM) significantly promoted ALP activity, mRNA levels of alkaline phosphatase biomineralization associated, collagen type 1, and runt‑related transcription factor‑2, protein levels of SMAD5, alkaline phosphatase and bone morphogenetic protein 2 protein expression and mineralized nodule formation in hPDLSCs. Furthermore, MAG significantly promoted the phosphorylation of SMAD2. Galunisertib inhibited the activation of SMAD2 and partially reversed the MAG‑mediated promotion of hPDLSC osteogenic differentiation. These data indicated that MAG promoted osteogenic differentiation of hPDLSCs potentially through TGF‑β/SMAD2 signaling. Therefore, MAG may help improve periodontal bone loss.

Introduction

Periodontal disease is a chronic inflammatory disease that is the leading cause of tooth loss in adults (1). This disease is characterized by an inflammatory response around the teeth, which is primarily caused by oral microbial biofilms and maintained by an uncoordinated immune inflammatory response, ultimately leading to progressive destruction of the tissues supporting the teeth (2). This condition is related to many systemic diseases, such as vascular disease, diabetes and heart disease (3). To the best of our knowledge, no suitable method for periodontal tissue regeneration has yet been developed.

The periodontal ligament contains pluripotent periodontal stem cells that express mesenchymal stem cell surface markers and show self-renewal and pluripotency (4). Periodontal ligament stem cells (PDLSCs) form cementum/periodontal ligament-like structures after transplantation in vivo, which indicates that PDLSCs play an important role in the reconstruction and regeneration of periodontal tissue, providing a new prospect for periodontal tissue regeneration (5). In addition, animal experiments have shown that PDLSCs from different sources (including human, canine, and porcine) can initiate the homing effect and promote the regeneration of periodontal tissue after implantation (6,7). Transplantation of PDLSCs effectively regenerates alveolar bone in alveolar defects in minipigs, showing encouraging results in preclinical trials (8,9). Due to their proliferative and pluripotent differentiation abilities, as well as their ability to form Sharpey fibers and cementum-like structures, PDLSCs are considered optimal seeding cells for periodontal engineering (10).

Researchers have increasingly examined new plant and fruit bioactive compounds that can be used to combat chronic disease and certain types of cancer (11,12). Mangiferin (MAG), a natural polyphenolic compound commonly found in mango and papaya, exhibits beneficial biological activities, including antioxidant, antitumor, antiviral, antidiabetic and immunomodulatory activities (1318). MAG has also been reported to have anti-osteoclast activity for the treatment and prevention of bone disease (19). To the best of our knowledge, however, the effect of MAG on the osteogenic differentiation of PDLSCs has not been reported.

The present study examined the effect of MAG on the proliferation and osteogenic differentiation of human PDLSCs (hPDLSCs) in vitro as well as the molecular pathways that are involved. It was hypothesized that MAG can promote the differentiation of hPDLSCs into osteoblasts and may be a potential drug for periodontal tissue regeneration.

Materials and methods

Primary culture of hPDLSCs

The first or second premolars that were removed from healthy individuals due to orthodontic needs were collected at the Beijing Stomatological Hospital after obtaining verbal patient consent, with approval of the Ethics Committee of Capital Medical University in China. A total of 10 patients (4 male, 6 females) aged 18–23 years were recruited from January 2021 to July 2021. Periodontal tissue was scraped from the middle third of the root of a healthy premolar extracted by orthodontic treatment. Tissues were minced and digested in equal volumes with collagenase type I (3 mg/ml) and neutral protease (4 mg/ml) for 1 h at 37°C. The isolated cells were then cultured in α-MEM with 10% fetal bovine serum and 1% penicillin/streptomycin (all Gibco) and placed in a 37°C and 5% CO2 cell incubator. Cells at third and fourth passage were used for the subsequent experiments.

Cell viability assay

The effect of MAG on viability of hPDLSCs was determined by Cell Counting Kit-8 (CCK-8) assays. Briefly, hPDLSCs were cultured in 96-well plates (1×104 cells/well) and cultured with 0, 50, 100, 200 or 500 µmol/l MAG (labeled MAG0, MAG50, MAG100, MAG200 or MAG500) for 7 days. Then, 10 µl CCK-8 reagent (Dojindo Laboratories, Inc.) was added dropwise to each well for 2 h. The optical density values in each well were measured at a wavelength of 490 nm under a microplate reader (Omega Bio-Tek, Inc.).

Osteogenic induction

The osteogenic medium consisted of 15% FBS (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin, 100 U/ml streptomycin, 1% glutamine (Gibco; Thermo Fisher Scientific, Inc.), 10 nmol/l dexamethasone (MilliporeSigma), 0.2 mmol/l ascorbic acid (MilliporeSigma) and 10 mmol/l sodium β-glycerophosphate (MilliporeSigma) in α-MEM (Gibco). MAG at 0, 200 and 500 µmol/l (labeled as MAG0, MAG200, MAG500) were selected for hPDLSC treatment in subsequent cell experiments.

Alkaline phosphatase (ALP) activity assay

ALP activity in hPDLSCs was assessed using an ALP activity assay kit (MilliporeSigma) and absorbance was measured at a wavelength of 520 nm on a microplate reader. ALP staining was performed using an ALP staining kit (Nanjing Jiancheng Bioengineering Institute) according to the manufacturer's protocol.

Alizarin red staining

Alizarin red staining was used to observe mineral deposition in each group. Cultured cells were fixed in 95% ethanol for 30 min and then stained with 0.1% Alizarin red staining solution (pH 4.2) for 20 min at room temperature. After cells were washed with PBS, 100 mmol/l cetylpyridinium chloride (MilliporeSigma) was added to each well and incubated at room temperature for 30 min. The absorbance was measured at a wavelength of 562 nm on a microplate reader.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) detection of osteogenesis-related genes

Total cellular RNA was extracted from hPDLSCs using TRIzol (Invitrogen, Thermo Fisher Scientific, Inc.) and a total of 2 µg RNA/sample was reverse transcribed to complementary DNA (cDNA) using a cDNA RT kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The expression of the osteogenesis-related genes ALP, biomineralization associated (ALPL), collagen type 1 (COL1) and runt-related transcription factor 2 (RUNX2) was determined by RT-qPCR using TaqMan Gene Expression Assay (Invitrogen, Thermo Fisher Scientific, Inc.), then qPCR was performed with denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec, 60°C for 30 sec and 72°C for 15 sec. The expression of the housekeeping gene GAPDH was used as an internal reference. Data were analyzed using the 2−∆∆Cq relative expression method (20). The primer sequences are listed in Table I.

Table I.

Reverse transcription-quantitative polymerase chain reaction primers used.

Table I.

Reverse transcription-quantitative polymerase chain reaction primers used.

Gene nameSequence, 5′→3′
GAPDH
  Forward CGGACCAATACGACCAAATCCG
  Reverse AGCCACATCGCTCAGACACC
ALPL
  Forward GACCTCCTCGGAAGACACTC
  Reverse TGAAGGGCTTCTTGTCTGTG
COL1
  Forward GCTGATGATGCCAATGTGGTT
  Reverse CCAGTCAGAGTGGCACATCTTG
RUNX2
  Forward GGAATGCCTCTGCTGTTATGAA
  Reverse GCTTCTGTCTGTGCCTTCTG

[i] ALPL, alkaline phosphate, biomineralization associated; COL1, collagen type 1; RUNX2, runt-related transcription factor 2.

Western blot analysis

Cells were harvested, washed and lysed in immunoprecipitation assay buffer (Beyotime Institute of Biotechnology). The protein content was determined using the BCA method. Proteins were separated by 12% (w/v) sodium dodecyl sulfate-polyacrylamide gel electrophoresis (20 µg protein per lane) and blotted onto polyvinylidene fluoride membranes. After blocking with Quick Blot Buffer (Applygen Technologies Inc.) at room temperature for 30 min, the proteins were detected by overnight incubation with rabbit polyclonal primary antibodies against RUNX2 (1:1,000, ab23981; Abcam), ALP (1:1,000, ab83295; Abcam), COL1 (1:1,000, ab233080, Abcam), HSP90 (1:1,000, ab13495; Abcam), TGF-β1 (1:1,000, ab92486; Abcam), p-SMAD2 (1:1,000, 3108T; Cell Signaling Technology), SMAD2 (1:1,000, 5339T; Cell Signaling Technology), SMAD3 (1:1,000, 9523T; Cell Signaling Technology) and β-actin (1:1,000, ab8227; Abcam) at 4°C. After washing, the membranes were incubated with horseradish peroxidase-conjugated goat-anti-rabbit IgG secondary antibody (1:2,000, cat. no. ab6721; Abcam) for 1 h at room temperature. Specific complexes were visualized using SuperEnhanced chemiluminescence detection kit (Applygen Technologies, Inc.). Band intensities were quantified using ImageJ 1.53 software (National Institutes of Health). The background was subtracted, and the signal of each target band was normalized to that of HSP90 or β-actin.

Galunisertib treatment of hPDLSCs

hPDLSCs were cultured for 5 days at 37°C in 4 groups as follows: untreated control, 200 µM MAG, 100 nM galunisertib or 100 nM galunisertib + 200 µM MAG. After multiple washes with PBS, hPDLSCs were harvested and subjected to western blot analysis and alizarin red staining analysis.

Statistical analysis

Statistical analysis was performed using SPSS 10.2 (SPSS, Inc.). One-way ANOVA and Tukey's post hoc test was used to assess differences between groups and P-values were calculated using unpaired Student's t test. All data are presented as the mean ± standard deviation (SD). A 95% confidence level (P<0.05) was considered to indicate a statistically significant difference. The number of replicates in each experiment is indicated in the figure legends.

Results

MAG has no effect on hPDLSC viability

Five concentrations of MAG (0, 50, 100, 200 and 500 µM) were selected for the experiments. The effect of different amounts of MAG on the viability of hPDLSCs was assessed using CCK-8 assays. The cell viability of hPDLSCs gradually increased from day 1 to 7, and different concentrations of MAG had no effect on the cell viability of hPDLSCs on days 1, 3, 5 and 7 compared with that of the MAG0 group (Fig. 1A). However, MAG500 significantly reduced cell viability compared with MAG50 and MAG10 at 7 days. Therefore, 0, 200 and 500 µM MAG were chosen for subsequent experiments.

MAG promotes osteogenic differentiation of hPDLSCs

ALP activity is important for bone mineralization and is a useful biochemical marker of bone formation (21). Both MAG200 and MAG500 significantly increased the ALP activity of hPDLSCs on day 4 of osteogenic induction (Fig. 1B). The ALP activity of the MAG200 group was the highest and was ~1.5 times that of the MAG0 group. The ALP activity of the MAG500 group was ~1.2 times that of the MAG0 group.

Next, the expression of osteogenic genes, including ALPL, COL1, and RUNX2, at 4 and 7 days was analyzed. MAG200 significantly increased the expression of ALPL on the 4th day (Fig. 1C). On both the 4 and 7th days, the expression of RUNX2 was significantly different from that of the MAG0 group. On day 7, neither MAG200 nor MAG500 had a significant effect on the expression of ALPL and COL1.

The protein levels of the osteogenesis-related genes COL1, RUNX2, SMAD5, ALP and bone morphogenetic protein 2 (BMP2) were semiquantified using western blotting (Fig. 2A and B). The SMAD5, ALP, and BMP2 protein levels in hPDLSCs in the MAG200 group were significantly upregulated after 7 days compared with the MAG0 group.

In addition to proteins associated with bone formation, mineralization is another marker for assessing osteogenesis (22). Calcium deposition was assessed to study the mineralization of cultured hPDLSCs in different groups. MAG treatment increased calcium nodules compared with the MAG0 group (Fig. 2C and D). However, the formation of mineralized nodules was not proportional to the concentration of MAG, with MAG200 resulting in the most mineralized nodules. As a confirmation of osteogenic induction, ALP staining performed after 4 days of culture showed positive staining for all of the MAG-containing groups (Fig. 2E). In particular, ALP activity was significantly upregulated in the MAG200 group compared with that in the MAG0 group. This was in accordance with the aforementioned ALP activity results. These results suggest that MAG served an important role in promoting mineralization and has a strong ability to induce osteogenic differentiation of hPDLSCs.

MAG promotes osteogenesis via the TGF-β/SMAD2 signaling pathway

To analyze the mechanism by which MAG promotes osteogenic differentiation, four key proteins of the TGF-β/SMAD2 signaling pathway, TGF-β, p-SMAD2, SMAD2 and SMAD3 were analyzed by western blotting. Galunisertib is a small molecule inhibitor of TGF-β receptor I kinase (23). The expression of TGF-β1 in the MAG200 group in the presence of galunisertib was significantly lower compared with the MAG200 group, indicating that galunisertib successfully inhibited expression of TGF-β1 (Fig. 3A and B). Western blotting showed that the expression levels of p-SMAD2, SMAD2 and SMAD3 were significantly upregulated in the MAG200 group after hPDLSCs were cultured for 7 days. Addition of the pathway inhibitor galunisertib partially reversed the upregulation of protein expression caused by MAG (Fig. 3A and B). The ratio of p-SMAD2/SMAD2 in the MAG200 + galunisertib group was significantly lower than the MAG200 group, indicating that galunisertib decreases SMAD2 phosphorylation (Fig. 3C). Alizarin red staining and calcium deposits quantification also demonstrated that the addition of galunisertib could partially inhibit the effect of MAG in promoting the osteogenic differentiation of hPDLSCs (Fig. 3D and E). Collectively, these data indicated that MAG activates the TGF-β/SMAD2 signaling pathway during osteogenic differentiation.

Discussion

Periodontal tissue is a complex group of tissues consisting of gingiva, periodontal ligament, cementum, and alveolar bone (24,25). Given this complexity, regeneration of lost or damage periodontal tissue due to periodontitis remains a challenge for current treatments (2628). PDLSCs can generate structures similar to the cementum/periodontal ligament in vivo, suggesting the important role of PDLSCs in periodontal regeneration (29). PDLSCs exhibit pluripotent differentiation and are good candidates for tissue engineering due to their ability to promote regeneration of dental and nondental tissues (30).

A number of previous studies have shown that MAG inhibits the inflammatory response (3133). Studies have shown that MAG has a notable inhibitory effect on intracellular adhesion molecule and endothelial leukocyte adhesion molecule expression, which are required for the transportation of leukocytes during inflammation (3436). Orally administered MAG (50 mg/kg body weight, once daily) was used to treat periodontitis in mice for 8 weeks; MAG was found to markedly inhibit alveolar bone loss, TNF-α production and NF-κB in gingival epithelial cells and phosphorylation of the JAK1-STAT1/3 pathway (37). Therefore, MAG has good therapeutic potential for prevention and treatment of periodontitis.

Researchers have found that MAG promotes bone tissue regeneration (3842). Sekiguchi et al (39) showed that MAG can promote osteoblastic bone formation by promoting cell proliferation and inducing cell differentiation through RUNX2 in pre-osteoblast MC3T3-E1 cells. Demeyer et al (40) prepared MAG-loaded chitosan-silica hybrid nanocomposite scaffolds using sol gel synthesis and freeze-drying processes; investigation of biomineralization and cell viability showed that the addition of bioactive MAG further promoted the effects of hybrid nanocomposite scaffolds in guided bone regeneration applications. Li et al (41) used a freeze-drying technique to prepare MAG-loaded poly(D, L-lactide-co-glycolide) scaffolds. The scaffolds were then implanted into the alveolar bone defect of diabetic rats and bone repair was examined using hematoxylin and eosin staining. Under diabetic conditions in vitro, the MAG-loaded scaffolds increased the histological score of bone regeneration and improved delayed alveolar bone defect healing in diabetic rats. Huh et al (42) isolated mesenchymal stem cells (MSCs) from the subchondral bone of rabbits and treated them with MAG and/or IL-1β. MAG induced chondrogenic differentiation of MSCs by upregulating the expression of several key chondrogenic markers, including TGF-β, BMP-2, and BMP-4. The experimental results of the present study are similar to those of the aforementioned experiments. In the present study, hPDLSCs were used as the research object, and MAG was found to promote the osteogenic differentiation of hPDLSCs. This result indicated that MAG may promote the regeneration of periodontal tissue by promoting the osteogenic differentiation of stem cells and may be a potential drug for periodontal treatment.

Osteoblast differentiation is critical for bone formation and involves the TGF-β/SMAD signaling pathway in bone morphogenesis (43). SMAD proteins are intermediate molecules that transmit the signal generated by the binding of TGF-β to its receptor from the cytoplasm to the nucleus, thereby playing an important role in signal transmission and regulation of transcription of downstream target genes (44). During signal transduction in the TGF-β/SMAD pathway, TGF-β first binds to its type II receptor and activates its type I receptor. Activated type I receptors promote the phosphorylation of SMAD2 or SMAD3 at the C-terminus, and these molecules bind with SMAD4 and translocate into the nucleus, thereby affecting osteoblast proliferation and differentiation (4547). Therefore, compounds or drugs that activate the SMAD signaling pathway through the TGF-β or BMP pathways can modulate osteoporosis (48).

Previous studies have shown that SMAD2/3 serves an important role in the regulation of bone formation (49) and the expression of SMAD2/3 and phosphorylated (p)-SMAD2/3 was found to be elevated during cementoblast differentiation and mineralization (50). Typically, activation of the TGF-β/SMAD signaling pathway mediates the phosphorylation of SMAD2/3, which dimerizes with SMAD4 and translocates to the nucleus, leading to transcription of downstream genes to direct cell differentiation (51). In the present study, western blot analysis showed that the addition of MAG could upregulate the expression of p-SMAD2, SMAD2, and SMAD3 in hPDLSCs, which revealed that MAG could activate the TGF-β/SMAD2 signaling pathway.

In the TGF-β/SMAD2 signaling pathway, serine in SMAD2 is directly phosphorylated by the TGF-β1 receptor, resulting in SMAD2 activation. Galunisertib (LY2157299 monohydrate) is a small molecule inhibitor of TGF-β receptor I kinase that specifically decreases SMAD2 phosphorylation and eliminates activation of the classic pathway (52). In the present study, addition of galunisertib partially reversed the MAG-mediated upregulation of the protein expression of p-SMAD2 and SMAD2. Alizarin red staining also indicated that galunisertib treatment could partially inhibit the effect of MAG in promoting the osteogenic differentiation of hPDLSCs. Collectively, these data indicated that MAG promotes the osteogenic differentiation of hPLDSCs by activating the TGF-β/SMAD2 signaling pathway.

In conclusion, MAG can promote osteogenic differentiation of hPDLSCs, and the TGF-β/SMAD2 signaling pathway was involved in this process. In addition, given the potential of MAG in antibacterial treatment and treatment of inflammation, studying the regulatory effect of MAG on bone regeneration has implications for the clinical treatment of periodontal disease. MAG may be an effective drug for preventing periodontitis and promoting periodontal bone regeneration.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82071144), National Key R&D Program of China (grant no. YFC1104304) and Young Scientist Program of Beijing Stomatological Hospital Capital Medical University (grant no. YSP202001).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YG and YB conceived the original idea and performed experiments. YG and LZ confirm the authenticity of all the raw data. LZ analyzed the data. YG, LZ and YB wrote the manuscript. YB supervised the project and give final approval of the version to be published. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The first or second premolars that were removed from healthy individuals due to orthodontic needs were collected at the Beijing Stomatological Hospital (Beijing, China) after obtaining patient verbal consent with approval of the Ethics Committee of Capital Medical University (approval no. KJ-2021-016-C-01-CS).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Highfield J: Diagnosis and classification of periodontal disease. Aust Dent J. 54 (Suppl 1):S11–S26. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Van Dyke TE: The management of inflammation in periodontal disease. J Periodontol. 79:1601–1608. 2008. View Article : Google Scholar : PubMed/NCBI

3 

John V, Alqallaf H and De Bedout T: Periodontal disease and systemic diseases: An update for the clinician. J Indiana Dent Assoc. 95:16–23. 2016.PubMed/NCBI

4 

Liu N, Gu B, Liu N, Nie X, Zhang B, Zhou X and Deng M: Wnt/β-catenin pathway regulates cementogenic differentiation of adipose tissue-deprived stem cells in dental follicle cell-conditioned medium. PLoS One. 9:e933642014. View Article : Google Scholar : PubMed/NCBI

5 

Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, Young M, Robey PG, Wang CY and Shi S: Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 364:149–155. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Hu L, Liu Y and Wang S: Stem cell-based tooth and periodontal regeneration. Oral Dis. 24:696–705. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Ouchi T and Nakagawa T: Mesenchymal stem cell-based tissue regeneration therapies for periodontitis. Regen Ther. 14:72–78. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Liu Y, Zheng Y, Ding G, Fang D, Zhang C, Bartold PM, Gronthos S, Shi S and Wang S: Periodontal ligament stem cell-mediated treatment for periodontitis in miniature swine. Stem Cells. 26:1065–1073. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Ding G, Liu Y, Wang W, Wei F, Liu D, Fan Z, An Y, Zhang C and Wang S: Allogeneic periodontal ligament stem cell therapy for periodontitis in swine. Stem Cells. 28:1829–1838. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Zhang Y, Xing Y, Jia L, Ji Y, Zhao B, Wen Y and Xu X: An in vitro comparative study of multisource derived human mesenchymal stem cells for bone tissue engineering. Stem Cells Dev. 27:1634–1645. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Scalbert A, Johnson IT and Saltmarsh M: Polyphenols: Antioxidants and beyond. Am J Clin Nutr. 81 (1 Suppl):215S–217S. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Mann S, Sarkar A, Sharma A, Gupta RK and Biswas S: Antitumor activity of choerospondias axillaris fruit extract by regulating the expression of SNCAIP and SNCA on MDA-MB-231 cells. Asian Pac J Cancer Prev. 23:1577–1586. 2022. View Article : Google Scholar : PubMed/NCBI

13 

Imran M, Arshad MS, Butt MS, Kwon JH, Arshad MU and Sultan MT: Mangiferin: A natural miracle bioactive compound against lifestyle related disorders. Lipids Health Dis. 16:842017. View Article : Google Scholar : PubMed/NCBI

14 

Wang M, Liang Y, Chen K, Wang M, Long X, Liu H, Sun Y and He B: The management of diabetes mellitus by mangiferin: Advances and prospects. Nanoscale. 14:2119–2135. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Walia V, Chaudhary SK and Kumar Sethiya N: Therapeutic potential of mangiferin in the treatment of various neuropsychiatric and neurodegenerative disorders. Neurochem Int. 143:1049392021. View Article : Google Scholar : PubMed/NCBI

16 

Morozkina SN, Nhung Vu TH, Generalova YE, Snetkov PP and Uspenskaya MV: Mangiferin as new potential anti-cancer agent and mangiferin-integrated polymer systems-a novel research direction. Biomolecules. 11:792021. View Article : Google Scholar : PubMed/NCBI

17 

Ren K, Li H, Zhou HF, Liang Y, Tong M, Chen L, Zheng XL and Zhao GJ: Mangiferin promotes macrophage cholesterol efflux and protects against atherosclerosis by augmenting the expression of ABCA1 and ABCG1. Aging (Albany NY). 11:10992–11009. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Zhu P, Liu C, Li B, Zhao C, Zhou T, Xue X and Zhang B: Mangiferin attenuates IL-1β-induced chondrocytes apoptosis. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 46:25–31. 2021.(In English, Chinese). PubMed/NCBI

19 

Ang E, Liu Q, Qi M, Liu HG, Yang X, Chen H, Zheng MH and Xu J: Mangiferin attenuates osteoclastogenesis, bone resorption, and RANKL-induced activation of NF-κB and ERK. J Cell Biochem. 112:89–97. 2011. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Rastogi D, Gautam N, Ara Z, Waliullah S and Srivastava RN: Prevalence of abnormal bone-specific alkaline phosphatase in orthopaedic trauma patients: A cross-sectional study from a tertiary trauma centre. Cureus. 14:e242642022.PubMed/NCBI

22 

Kato K, Ozaki M, Nakai K, Nagasaki M, Nakajima J, Koshi R, Tanaka H, Kawato T and Tonogi M: Effect of azithromycin on mineralized nodule formation in MC3T3-E1 cells. Curr Issues Mol Biol. 43:1451–1459. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Hira SK, Rej A, Paladhi A, Singh R, Saha J, Mondal I, Bhattacharyya S and Manna PP: Galunisertib drives treg fragility and promotes dendritic cell-mediated immunity against experimental lymphoma. iScience. 23:1016232020. View Article : Google Scholar : PubMed/NCBI

24 

Hassell TM: Tissues and cells of the periodontium. Periodontol. 2000.3:9–38. 1993. View Article : Google Scholar

25 

Cho MI and Garant PR: Development and general structure of the periodontium. Periodontol. 2000.24:9–27. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Rasperini G, Tavelli L, Barootchi S, McGuire MK, Zucchelli G, Pagni G, Stefanini M, Wang HL and Giannobile WV: Interproximal attachment gain: The challenge of periodontal regeneration. J Periodontol. 92:931–946. 2021. View Article : Google Scholar : PubMed/NCBI

27 

de Jong T, Bakker AD, Everts V and Smit TH: The intricate anatomy of the periodontal ligament and its development: Lessons for periodontal regeneration. J Periodontal Res. 52:965–974. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Hughes FJ, Ghuman M and Talal A: Periodontal regeneration: A challenge for the tissue engineer? Proc Inst Mech Eng H. 224:1345–1358. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Tomokiyo A, Wada N and Maeda H: Periodontal ligament stem cells: Regenerative potency in periodontium. Stem Cells Dev. 28:974–985. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Li Y, Liu A, Zhang L, Wang Z, Hui N, Zhai Q, Zhang L, Jin Z and Jin F: Epithelial cell rests of malassez provide a favorable microenvironment for ameliorating the impaired osteogenic potential of human periodontal ligament stem cells. Front Physiol. 12:7352342021. View Article : Google Scholar : PubMed/NCBI

31 

Wang X, Yuwen T and Yanqin T: Mangiferin inhibits inflammation and cell proliferation, and activates proapoptotic events via NF-κB inhibition in DMBA-induced mammary carcinogenesis in rats. J Environ Pathol Toxicol Oncol. 40:1–9. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Dong M, Li L, Li G, Song J, Liu B, Liu X and Wang M: Mangiferin protects against alcoholic liver injury via suppression of inflammation-induced adipose hyperlipolysis. Food Funct. 11:8837–8851. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Bulugonda RK, Kumar KA, Gangappa D, Beeda H, Philip GH, Muralidhara Rao D and Faisal SM: Mangiferin from Pueraria tuberosa reduces inflammation via inactivation of NLRP3 inflammasome. Sci Rep. 7:426832017. View Article : Google Scholar : PubMed/NCBI

34 

Beltrán Núñez AE, Naranjo NL, Penabad CR, Sironi M, Rodríguez GQ, Garrido GG and Hernández RD: VIMANG® y mangiferina inhiben la expresión de ICAM-1 en células endoteliales estimuladas con citocinas proinflamatorias. Rev Cubana Invest Biomed. 22:164–172. 2003.

35 

Yang H, Bai W, Gao L, Jiang J, Tang Y, Niu Y, Lin H and Li L: Mangiferin alleviates hypertension induced by hyperuricemia via increasing nitric oxide releases. J Pharmacol Sci. 137:154–161. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Dou W, Zhang J, Ren G, Ding L, Sun A, Deng C, Wu X, Wei X, Mani S and Wang Z: Mangiferin attenuates the symptoms of dextran sulfate sodium-induced colitis in mice via NF-κB and MAPK signaling inactivation. Int Immunopharmacol. 23:170–178. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Li H, Wang Q, Ding Y, Bao C and Li W: Mangiferin ameliorates porphyromonas gingivalis-induced experimental periodontitis by inhibiting phosphorylation of nuclear factor-κB and Janus kinase 1-signal transducer and activator of transcription signaling pathways. J Periodontal Res. 52:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Bai Y, Liu C, Fu L, Gong X, Dou C, Cao Z, Quan H, Li J, Kang F, Dai J, et al: Mangiferin enhances endochondral ossification-based bone repair in massive bone defect by inducing autophagy through activating AMP-activated protein kinase signaling pathway. FASEB J. 32:4573–4584. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Sekiguchi Y, Mano H, Nakatani S, Shimizu J, Kataoka A, Ogura K, Kimira Y, Ebata M and Wada M: Mangiferin positively regulates osteoblast differentiation and suppresses osteoclast differentiation. Mol Med Rep. 16:1328–1332. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Demeyer S, Athipornchai A, Pabunrueang P and Trakulsujaritchok T: Development of mangiferin loaded chitosan-silica hybrid scaffolds: Physicochemical and bioactivity characterization. Carbohydr Polym. 261:1179052021. View Article : Google Scholar : PubMed/NCBI

41 

Li H, Liao H, Bao C, Xiao Y and Wang Q: Preparation and evaluations of mangiferin-loaded PLGA scaffolds for alveolar bone repair treatment under the diabetic condition. AAPS PharmSciTech. 18:529–538. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Huh JE, Koh PS, Seo BK, Park YC, Baek YH, Lee JD and Park DS: Mangiferin reduces the inhibition of chondrogenic differentiation by IL-1β in mesenchymal stem cells from subchondral bone and targets multiple aspects of the Smad and SOX9 pathways. Int J Mol Sci. 15:16025–16042. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Yu J, Xu L, Li K, Xie N, Xi Y, Wang Y, Zheng X, Chen X, Wang M and Ye X: Zinc-modified calcium silicate coatings promote osteogenic differentiation through TGF-β/Smad pathway and osseointegration in osteopenic rabbits. Sci Rep. 7:34402017. View Article : Google Scholar : PubMed/NCBI

44 

Luo K: Signaling CROSS TALK BETWeen TGF-β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol. 9:a0221372017. View Article : Google Scholar : PubMed/NCBI

45 

Runyan CE, Liu Z and Schnaper HW: Phosphatidylinositol 3-kinase and Rab5 GTPase inversely regulate the Smad anchor for receptor activation (SARA) protein independently of transforming growth factor-β1. J Biol Chem. 287:35815–35824. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Ota K, Quint P, Ruan M, Pederson L, Westendorf JJ, Khosla S and Oursler MJ: TGF-β induces Wnt10b in osteoclasts from female mice to enhance coupling to osteoblasts. Endocrinology. 154:3745–3752. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Li XL, Liu YB, Ma EG, Shen WX, Li H and Zhang YN: Synergistic effect of BMP9 and TGF-β in the proliferation and differentiation of osteoblasts. Genet Mol Res. 14:7605–7615. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Wang W, Rigueur D and Lyons KM: TGFβ as a gatekeeper of BMP action in the developing growth plate. Bone. 137:1154392020. View Article : Google Scholar : PubMed/NCBI

49 

Heo SY, Ko SC, Nam SY, Oh J, Kim YM, Kim JI and Jung WK: Fish bone peptide promotes osteogenic differentiation of MC3T3-E1 pre-osteoblasts through upregulation of MAPKs and Smad pathways activated BMP-2 receptor. Cell Biochem Funct. 36:137–146. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Li L, Zhu Z, Xiao W and Li L: Multi-walled carbon nanotubes promote cementoblast differentiation and mineralization through the TGF-β/Smad signaling pathway. Int J Mol Sci. 16:3188–3201. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Whitman M: Smads and early developmental signaling by the TGFbeta superfamily. Genes Dev. 12:2445–2462. 1998. View Article : Google Scholar : PubMed/NCBI

52 

Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, et al: Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther. 9:4479–4499. 2015.PubMed/NCBI

Related Articles

Journal Cover

August-2022
Volume 26 Issue 2

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
Gu Y, Zhang L and Bai Y: Mangiferin promotes the osteogenic differentiation of human periodontal ligament stem cells via TGF‑β/SMAD2 signaling. Mol Med Rep 26: 266, 2022
APA
Gu, Y., Zhang, L., & Bai, Y. (2022). Mangiferin promotes the osteogenic differentiation of human periodontal ligament stem cells via TGF‑β/SMAD2 signaling. Molecular Medicine Reports, 26, 266. https://doi.org/10.3892/mmr.2022.12782
MLA
Gu, Y., Zhang, L., Bai, Y."Mangiferin promotes the osteogenic differentiation of human periodontal ligament stem cells via TGF‑β/SMAD2 signaling". Molecular Medicine Reports 26.2 (2022): 266.
Chicago
Gu, Y., Zhang, L., Bai, Y."Mangiferin promotes the osteogenic differentiation of human periodontal ligament stem cells via TGF‑β/SMAD2 signaling". Molecular Medicine Reports 26, no. 2 (2022): 266. https://doi.org/10.3892/mmr.2022.12782