Open Access

Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling

  • Authors:
    • Ping Yang
    • Fenghua Shi
    • Yanli Zhang
  • View Affiliations

  • Published online on: December 22, 2022     https://doi.org/10.3892/etm.2022.11773
  • Article Number: 74
  • Copyright: © Yang 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

Periodontitis is the chronic inflammation of the periodontal tissue. The present study aimed to investigate the role of baricitinib, a Janus kinase (JAK)1/2 inhibitor, in periodontitis by using a lipopolysaccharide (LPS)‑induced human periodontal ligament stem cell (PDLSC) model. The viability of PDLSCs stimulated by LPS was assessed in the presence of baricitinib by Cell Counting Kit‑8 assay. The induction of oxidative stress was evaluated by detecting the intracellular reactive oxygen species (ROS) levels, superoxide dismutase (SOD) activity and glutathione (GSH) content. ELISA and reverse transcription‑quantitative PCR were used to determine the levels of inflammatory factors TNF‑α, IL‑1β and IL‑6. Alkaline phosphatase (ALP) activity and alizarin red staining were used to assess the osteogenic differentiation of PDLSCs. The expression levels of osteogenic differentiation‑ and JAK/signal transducer and activator of transcription (STAT) signaling‑associated proteins were estimated with western blotting. RO8191, an agonist of the JAK/STAT pathway, was used to treat PDLSCs to investigate the regulatory mechanism of baricitinib. The results indicated that baricitinib elevated the LPS‑induced decrease in cell viability. LPS‑triggered oxidative stress and inflammation were inhibited by baricitinib, as demonstrated by the decreased levels of ROS, TNF‑α, IL‑1β, IL‑6 and increased levels of SOD and GSH. In addition, baricitinib caused a marked elevation in ALP activity and mineralization ability of PDLSCs, as determined by the upregulated osteocalcin and Runt‑related transcription factor 2 expression. Moreover, the expression levels of phosphorylated (p)‑JAK1, p‑JAK2 and p‑STAT3 were downregulated by baricitinib in a dose‑dependent manner. Furthermore, addition of RO8191 restored the effect of baricitinib on the induction of oxidative stress, inflammation and osteogenic differentiation of PDLSCs exposed to LPS. Collectively, these findings suggested that baricitinib alleviated oxidative stress and inflammation and promoted osteogenic differentiation of LPS‑induced PDLSCs by inhibiting JAK/STAT signaling.

Introduction

Periodontitis, an infectious human chronic inflammatory disease of which the age-standardized prevalence rate increased by 8.44% (6.62-10.59%) worldwide from 1990 to 2019(1), is caused by accumulation of subgingival plaque and the action of gram-negative anaerobic bacteria, such as Porphyromonas gingivalis and Treponema denticola (2,3). It is characterized by the progressive destruction of tooth-supporting tissue, eventually leading to tooth loss (4,5). A number of studies have demonstrated that periodontitis is associated with the initiation or progression of various systemic diseases, such as cardiovascular and chronic kidney disease and diabetes (6-8). Although progress has been achieved in the treatment of periodontitis, including skin flap curettage, scaling and root planting, the effects of the aforementioned therapies only control the development of this condition (9-11). Therefore, it is of clinical importance to investigate promising therapeutic agents for periodontitis to improve oral health and avert the development of systemic disease.

Periodontal ligament stem cells (PDLSCs), mesenchymal stem cells located in the periodontal ligament tissue, harbor osteogenic potential and regenerative capacity to recover lost/damaged periodontal tissue (12,13). PDLSCs have been shown to migrate to the site of periodontal disease and mediate periodontal regeneration (14,15). PDLSCs are considered an ideal cell source for periodontal tissue regeneration and repair (16,17). A body of evidence indicates that endotoxins generated from periodontal pathogens, notably lipopolysaccharide (LPS) produced by Porphyromonas gingivalis, disrupt microenvironment homeostasis and damage periodontal tissue by inhibiting the viability of PDLSCs and causing disturbance of periodontal ligament cell differentiation (18,19). LPS is a potent stimulator of inflammation that produces proinflammatory cytokines. This compound has been widely used for construction of in vitro experimental models of periodontitis (20,21). It is crucial to control the inflammatory damage of PDLSCs for the restoration of the tissues and to inhibit progression of periodontitis.

Baricitinib is an oral drug commonly used in treatment of rheumatoid arthritis (22). A recent clinical study has suggested that baricitinib improves the periodontal health of patients with rheumatoid arthritis and decrease the inflammatory response in periodontal tissue (23). Baricitinib suppresses osteoclastogenesis by downregulating receptor activator of nuclear factor-κB ligand (RANKL) expression in osteoblasts (24). In addition, as a Janus kinase (JAK)1/2 inhibitor, baricitinib increases bone mass in physiological conditions and ameliorates pathological bone loss by stimulating osteoblast function (25). Meanwhile, the abnormal expression of STAT3, an important member of STAT protein family, has been reported to mediate the over-activation of JAK-STAT3 signaling pathway, which is related to the process of periodontitis (26). However, whether baricitinib affects inflammation and osteogenic differentiation of LPS-induced PDLSCs and its potential regulatory mechanisms remains to be elucidated.

Materials and methods

Cell culture

Human PDLSCs provided by Shanghai Chuntest Biotechnology Co., Ltd. were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) with 10% fetal bovine serum (FBS; HyClone; Cytiva) at 37˚C in an incubator with 5% CO2. To induce osteogenic differentiation, PDLSCs were cultured in osteogenic differentiation medium, which was composed of α-MEM (Sigma-Aldrich; Merck KGaA) containing 10 mM β-glycerophosphate, 10% FBS, 50 mM ascorbic acid and 10 nM dexamethasone for 14 days in a 37˚C incubator with 5% CO2, as previously described (27).

Treatment protocol

LPS (10 µg/ml; Sigma-Aldrich; Merck KGaA) was used to induce PDLSCs for 24 h at 37˚C to simulate the inflammatory microenvironment of periodontitis, as previously described (21). The biological source of LPS was Porphyromonas gingivalis. For baricitinib-treated groups, PDLSCs were treated with baricitinib (1.0, 2.5 and 5.0 µM; ChemScene) for 24 h at 37˚C in the presence or absence of LPS (24). In addition, PDLSCs were pretreated with RO8191 (20 µM; MedChemExpress), an agonist of the JAK/signal transducer and activator of transcription (STAT) pathway, for 1 h at 37˚C to investigate the regulatory effect of baricitinib on this signaling pathway, as previously described (28).

Cell viability assay

The viability of PDLSCs was evaluated by Cell Counting Kit-8 (CCK-8) assay (Shanghai Yeasen Biotechnology Co., Ltd.) A total of 5x103 PDLSCs were seeded in a 96-well plate. LPS or baricitinib was employed to treat PDLSCs 24 h prior to addition of CCK-8 solution (10 µl). Following incubation for another 4 h, a microplate reader (Molecular Devices, LLC) was applied to record the optical density values at 450 nm.

Assessment of intracellular reactive oxygen species (ROS)

The 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA) probe is a commonly used probe for detecting intracellular ROS (29). PDLSCs were seeded in 96-well plates at a density of 2x104 cells/well. Following treatment with LPS, baricitinib or RO8191 and PDLSCs were incubated with 10 µM DCFH-DA (Beyotime Institute of Biotechnology) for 20 min at 37˚C. An inverted fluorescence microscope (Olympus Corporation) (magnification, x400) was used to measure the fluorescence intensity corresponding to ROS levels from three random fields.

Determination of superoxide dismutase (SOD) activity and glutathione (GSH) content

The activity of SOD and content of GSH in the cell supernatant was evaluated by the SOD assay kit (cat. no. A001-3-2) and GSH assay kit (cat. no. A006-2-1) supplied by Nanjing Jiancheng Bioengineering Institute according to the manufacturer's protocol. The absorbance was detected using a microplate reader (Molecular Devices, LLC) at a wavelength of 450 and 420 nm.

Detection of inflammatory factors

The protein levels of TNF-α, IL-1β and IL-6 in cell supernatant was assessed by human TNF-α ELISA kit (cat. no. F02810), human IL-1β ELISA kit (cat. no. F01220) and human IL-6 ELISA kit (cat. no. F01310) (Shanghai Xitang Biotechnology Co., Ltd.) according to the manufacturer's instructions. The optical density values were determined at 450 nm by a microplate reader (Molecular Devices, LLC).

Assessment of alkaline phosphatase (ALP) activity

Following incubation at 37˚C in osteogenic medium for 14 days, ALP activity of the PDLSCs was assessed by an ALP assay kit (cat. no. A059-2-1; Nanjing Jiancheng Biotechnology Institute) according to the manufacturer's instructions. The optical density was detected at 520 nm using a microplate reader (Molecular Devices, LLC).

Alizarin red staining

The mineralization potential of PDLSCs was determined using alizarin red staining after cells were cultured at 37˚C seeded into a 24-well plate at a density of 250 cells/well in the osteogenic medium with or without LPS, baricitinib or RO8191 for 14 days. PDLSCs were incubated with 4% paraformaldehyde for 30 min at 4˚C, followed by staining with 1% alizarin red solution (Sigma-Aldrich; Merck KGaA) for 15 min at 37˚C. Following washing three times with PBS, the images were captured by a light microscope (magnification, x200; Olympus Corporation) with three fields for each group. To quantify mineralization, calcium deposits were desorbed using 10% cetylpyridinium chloride (Sigma-Aldrich; Merck KGaA) for 30 min at room temperature and the optical density value was detected using a plate reader at 562 nm (Molecular Devices, LLC).

Reverse transcription-quantitative PCR

Total RNA was isolated from PDLSCs with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). RT was performed to generate complementary DNA by PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd.) according to the manufacturer's instructions. The detection of mRNA expression was performed in the ABI 7500 system (Applied Biosystems; Thermo Fisher Scientific, Inc.) using the QuantiNova SYBR Green PCR kit (Qiagen GmbH). The thermocycling conditions were as follows: Initial denaturation at 95˚C for 5 min, followed by 40 cycles of denaturation for 15 sec at 95˚C and annealing for 30 sec at 60˚C. The primer sequences were as follows: TNF-α forward, 5'-CCTCTCTCTAATCAGCCCTCTG-3' and reverse, 5'-GAGGACCTGGGAGTAGATGAG-3'; IL-1β forward, 5'-ATGATGGCTTATTACAGTGGCAA-3' and reverse, 5'-GTCGGAGATTCGTAGCTGGA-3'; IL-6 forward, 5'-ACTCACCTCTTCAGAACGAATTG-3' and reverse, 5'-CCATCTTTGGAAGGTTCAGGTTG-3' and GAPDH forward, 5'-GGAGCGAGATCCCTCCAAAAT-3' and reverse, 5'-GGCTGTTGTCATACTTCTCATGG-3'. The calculation of relative mRNA expression was performed with the 2-ΔΔCq method (30). Primers were synthesized by Shanghai GenePharma Co., Ltd. GAPDH was utilized as an endogenous control.

Western blot analysis

PDLSCs were lysed in RIPA lysis buffer (Beyotime Institute of Biotechnology) to extract the total protein, which was quantified using a bicinchoninic protein assay kit (Beyotime Institute of Biotechnology). Protein separation was performed using 10% SDS-PAGE (30 µg/lane). The separated proteins were transferred to PVDF membranes. The blots were blocked with 5% non-fat dry milk at room temperature for 2 h, followed by incubation with primary antibodies including p-JAK1 (1:1,000; cat. no. ab138005; Abcam), JAK1 (1:1,000; cat. no. ab133666; Abcam), p-JAK2 (1:1,000; cat. no. ab32101; Abcam), JAK2 (1:5,000; cat. no. ab108596; Abcam), p-STAT3 (1:1,000; cat. no. ab68153; Abcam), STAT3 (1:1,000; cat. no. ab267373; Abcam), OCN (1:1,000; cat. no. ab133612; Abcam), RUNX2 (1:1,000; cat. no. ab236639; Abcam) and GAPDH (1:2,500; cat. no. ab9485; Abcam) overnight at 4˚C. Subsequently, membranes were labeled with a horseradish peroxidase-conjugated secondary antibody (1:5,000; cat. no. ab205718; Abcam) for 1 h at room temperature. The bands were visualized using an ECL kit (Beyotime Institute of Biotechnology) and detection system (MilliporeSigma). The band densities were evaluated using ImageJ software (v1.8.0; National Institutes of Health).

Statistical analysis

All experiments were repeated independently in triplicate and data are expressed as the mean ± standard deviation. GraphPad Prism 6.0 (GraphPad Software, Inc.) was used for data analysis. The level of significance was analyzed by one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Baricitinib increases viability of PDLSCs exposed to LPS

Firstly, cell viability was evaluated by CCK-8 assay following treatment of PDLSCs with baricitinib (1.0, 2.5 and 5.0 µM). Baricitinib treatment exhibited no significant effect on the viability of PDLSCs compared with the control group (Fig. 1A). PDLSCs were stimulated by LPS with or without baricitinib. LPS treatment led to a significant decrease in PDLSC viability compared with the control group (Fig. 1B). Baricitinib dose-dependently increased the viability of PDLSCs following LPS treatment and PDLSCs viability was significantly exacerbated when treated by 5 µM baricitinib upon exposure to LPS. These results demonstrated that baricitinib reversed the LPS-induced decrease in viability of PDLSCs.

Baricitinib alleviates LPS-induced oxidative stress and inflammation in PDLSCs

The levels of intracellular ROS were evaluated using DCFH-DA as a probe; fluorescence intensity reflected the content of ROS. The fluorescence intensity in the LPS group was markedly enhanced compared with that of the control group (Fig. 2A). However, baricitinib decreased the fluorescence intensity induced by LPS in a concentration-dependent manner. Moreover, decreased SOD activity and GSH content caused by the LPS challenge gradually recovered following baricitinib treatment (Fig. 2B and C). In addition, levels of TNF-α, IL-1β, and IL-6 were elevated following incubation of PDLSCs with LPS (Fig. 2D-F). Additional treatment with baricitinib reversed the impact of LPS on expression levels of the aforementioned inflammatory factors in a concentration-dependent manner. Consistently, elevated mRNA expression levels of these inflammatory factors (TNF-α, IL-1β and IL-6) induced by LPS were also decreased following baricitinib treatment (Fig. 2G). These data provide evidence that baricitinib alleviated LPS-induced oxidative stress and inflammation in PDLSCs.

Baricitinib promotes osteogenic differentiation in LPS-stimulated PDLSCs

The effect of baricitinib on osteogenic differentiation of PDLSCs challenged with LPS was investigated. ALP activity was significantly decreased in LPS group compared with that of the control group (Fig. 3A). By contrast, baricitinib dose-dependently increased ALP activity relative to the LPS group. The results of alizarin red staining indicated that the mineralization degree of PDLSCs was enhanced in the control group; this effect was reduced following stimulation of the cells with LPS (Fig. 3B and C). Furthermore, in the presence of baricitinib, the mineralization ability of PDLSCs increased in a dose-dependent manner. Moreover, the expression levels of proteins related to osteogenic differentiation, including osteocalcin (OCN) and Runt-related transcription factor (Runx) 2, were downregulated following treatment with LPS compared with the control group, as determined by western blot analysis. This effect was reversed and the levels of OCN and Runx2 were upregulated following subsequent treatment of the cells with baricitinib (Fig. 3D). These observations revealed that baricitinib promoted the osteogenic differentiation of PDLSCs stimulated by LPS.

Baricitinib inactivates the JAK/STAT signaling pathway in LPS-stimulated PDLSCs

To investigate the underlying mechanism by which baricitinib regulates the process of PDLSC injury and osteogenic differentiation triggered by LPS, expression of specific proteins involved in the JAK/STAT pathway was assessed using western blot analysis. The expression levels of phosphorylated (p)-JAK1, p-JAK2 and p-STAT3 were significantly increased in the LPS group compared with the control group (Fig. 4). By contrast, baricitinib caused a gradual downregulation in the expression levels of these proteins in a dose-dependent manner; the highest inhibitory effect was noted in the 5 µM-treated group. Thereafter, 5 µM baricitinib was applied to the subsequent experiments. Collectively, these data suggested that baricitinib suppressed the JAK/STAT signaling pathway in LPS-induced PDLSCs.

Baricitinib improves LPS-induced oxidative stress and inflammation in PDLSCs by inhibiting JAK/STAT signaling

To clarify the regulatory effect of baricitinib on the JAK/STAT signaling pathway in LPS-stimulated PDLSCs, RO8191, an agonist of the JAK/STAT pathway, was used to treat PDLSCs. RO8191 addition significantly upregulated p-JAK1, p-JAK2 and p-STAT3 expression in PDLSCs treated with LPS and baricitinib compared with the LPS +Baricitinib group (Fig. 5). Subsequently, levels of oxidative stress and inflammation were evaluated. RO8191 significantly increased ROS levels compared with those noted in the LPS + baricitinib group, as determined by the decreased SOD activity and GSH content (Fig. 6A-C). Moreover, the decline in the TNF-α, IL-1β and IL-6 levels induced by baricitinib was significantly reversed following RO8191 treatment (Fig. 6D-G). These findings confirmed that baricitinib exerted antioxidant and anti-inflammatory effects in LPS-stimulated PDLSCs by inhibiting JAK/STAT signaling.

Baricitinib promotes osteogenic differentiation of PDLSCs challenged with LPS by inhibiting JAK/STAT signaling

Subsequently, osteogenic differentiation of LPS-stimulated PDLSCs was assessed with or without baricitinib or RO8191. The results indicated that addition of RO8191 led to a significant decrease in ALP activity compared with that in the LPS + baricitinib group (Fig. 7A). Alizarin red staining suggested that the mineralization degree of PDLSCs was significantly decreased following addition of RO8191 in PDLSCs co-treated with LPS and baricitinib (Fig. 7B and C). Concomitantly, significant downregulation of OCN and Runx2 expression was observed in the LPS + baricitinib + RO8191 group compared with the LPS + baricitinib group (Fig. 7D). In summary, these results revealed that baricitinib promoted osteogenic differentiation of PDLSCs exposed to LPS by inhibiting JAK/STAT signaling.

Discussion

Periodontitis is a non-infectious chronic inflammatory disease that is primarily induced by microbial attack and affects tooth support structures. This condition is prevalent worldwide and affects large populations with age-standardized prevalence rate increased by 8.44% worldwide from 1990 to 2019 (1,31,32). Due to excellent osteogenic capacity, PDLSCs serve a crucial role in maintaining periodontal bone mass (33). However, the inflammatory microenvironment suppresses the osteogenic capacity of PDLSCs, which is also an important pathogenic feature of periodontitis (34). Therefore, it is critical to discover a novel drug that protects PDLSCs from inflammation-associated injury and improves their functional characteristics. The present study demonstrated the effects of baricitinib on LPS-induced damage of PDLSCs and the decrease of osteogenic differentiation.

Periodontitis is an inflammatory disease mediated by immunization; LPS produced by dominant bacteria in gingival crevices serves an indispensable role in the occurrence and progression of this condition (35). Exposure to LPS leads to increased levels of certain inflammatory factors, including TNF-α, IL-1β and IL-6, in PDLSCs (36). Numerous studies have shown that LPS induces PDLSCs to produce and accumulate a large amount of ROS, which disrupts the balance between oxidative and antioxidant systems, resulting in cell and tissue damage (37,38). SOD and GSH are considered specific parameters of oxidative stress. A significant increase in the levels of ROS and expression levels of TNF-α, IL-1β and IL-6, and a decrease in the levels of SOD and GSH, were noted in PDLSCs following stimulation with LPS, which was consistent with previous studies (36-38). As a U.S Food and Drug Administration-approved oral JAK1/2 inhibitor, baricitinib has been used to treat moderate to extreme rheumatoid arthritis (39). A clinical study demonstrated that clinical manifestations and inflammatory biomarkers were notably improved in patients with autoinflammatory interferonopathy following baricitinib treatment (40). Baricitinib is reported to inhibit tetradecanoylphorbol-13-acetate-induced psoriasis and skin inflammation in a mouse model (41). Baricitinib improves the periodontal status of patients with rheumatoid arthritis and decreases the inflammatory response in the periodontal tissue (23). In the present study, the antioxidant and anti-inflammatory effects of baricitinib were verified in LPS-induced PDLSCs, suggesting the potential application of this compound in the treatment of periodontitis.

Subsequently, the effects of baricitinib on osteogenic differentiation of PDLSCs incubated with LPS were analyzed by evaluating the expression of several osteogenic differentiation markers and mineralization in PDLSCs. ALP is an early marker of osteogenic differentiation and a key hydrolase in cellular osteogenic differentiation (42). The increase in the activity of ALP reflects maturation of osteogenic differentiation (43). Alizarin red staining forms mineralized nodules through the specific binding of alizarin red S to calcium ions, which indicates mineralization capacity of cells (44). OCN, a non-collagen protein abundant in the bone tissue, is a specific indicator of bone metabolism and osteocyte activity; its expression levels reflect the rate of bone formation (45). Runx2 is a key gene of bone formation and an important indicator of osteogenic differentiation of mesenchymal stem cells (46). Baricitinib can increase bone mass in steady-state conditions and ameliorate pathological bone loss by stimulating osteoblast function (25). Murakami et al (24) demonstrated that baricitinib inhibits osteoclast formation in vitro by suppressing the expression of RANKL. The present results indicated that under the treatment of baricitinib, levels of ALP, Runx2 and OCN and mineralization degree were elevated, which confirmed that baricitinib promoted osteogenic differentiation of PDLSCs, highlighting its potential to treat periodontitis.

To investigate the underlying mechanism of baricitinib on the impact of LPS-stimulated PDLSCs, expression levels of proteins involved in the JAK/STAT pathway were assessed. A previous study revealed that Porphyromonas gingivalis activates the JAK/STAT signaling pathway and enhances ROS accumulation, thereby promoting the occurrence and development of periodontitis (47). Downregulation of Toll-like receptor 4 expression and the JAK1/STAT3 signaling pathway can improve diabetic periodontitis in a mouse model (48). Zheng et al (26) revealed that inhibition of JAK/STAT signaling suppresses progression of chronic periodontitis. Here, upregulation of p-JAK1, JAK2 and STAT3 expression in PDLSCs induced by LPS was inhibited by baricitinib. Subsequent addition of RO8191, an agonist of the JAK/STAT pathway, abolished the protective effects of baricitinib on LPS-triggered oxidative stress, inflammation and loss of osteogenic differentiation, suggesting that this compound alleviated LPS-induced PDLSC injury and promoted osteogenic differentiation by inhibiting JAK/STAT signaling.

The present study demonstrated that baricitinib, with its antioxidant, anti-inflammatory and pro-osteogenic differentiation effects, may be a promising agent in protecting PDLSCs from periodontitis-induced injury. The present study may provide insight into the use of baricitinib for the treatment of periodontitis. Further in vivo animal studies will be performed in the future to confirm the effects of baricitinib on periodontitis.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

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

PY and FS designed the study. PY, FS and YZ performed the experiments and analyzed the data. PY drafted the manuscript and interpreted the data. YZ revised the manuscript for important intellectual content. All authors have read and approved the final manuscript. PY and YZ confirm the authenticity of all the raw data.

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 

Chen MX, Zhong YJ, Dong QQ, Wong HM and Wen YF: Global, regional, and national burden of severe periodontitis, 1990-2019: An analysis of the global burden of disease study 2019. J Clin Periodontol. 48:1165–1188. 2021.PubMed/NCBI View Article : Google Scholar

2 

Socransky SS, Haffajee AD, Cugini MA, Smith C and Kent RL Jr: Microbial complexes in subgingival plaque. J Clin Periodontol. 25:134–144. 1998.PubMed/NCBI View Article : Google Scholar

3 

Byrne SJ, Dashper SG, Darby IB, Adams GG, Hoffmann B and Reynolds EC: Progression of chronic periodontitis can be predicted by the levels of Porphyromonas gingivalis and Treponema denticola in subgingival plaque. Oral Microbiol Immunol. 24:469–477. 2009.PubMed/NCBI View Article : Google Scholar

4 

Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, Flemmig TF, Garcia R, Giannobile WV, Graziani F, et al: Periodontitis: Consensus report of workgroup 2 of the 2017 world workshop on the classification of periodontal and peri-implant diseases and conditions. J Periodontol. 89 (Suppl 1):S173–S182. 2018.PubMed/NCBI View Article : Google Scholar

5 

Hajishengallis G: Periodontitis: From microbial immune subversion to systemic inflammation. Nat Rev Immunol. 15:30–44. 2015.PubMed/NCBI View Article : Google Scholar

6 

Polak D and Shapira L: An update on the evidence for pathogenic mechanisms that may link periodontitis and diabetes. J Clin Periodontol. 45:150–166. 2018.PubMed/NCBI View Article : Google Scholar

7 

Wallace K, Shafique S and Piamjariyakul U: The relationship between oral health and hemodialysis treatment among adults with chronic kidney disease: A systematic review. Nephrol Nurs J. 46:375–394. 2019.PubMed/NCBI

8 

Khumaedi AI, Purnamasari D, Wijaya IP and Soeroso Y: The relationship of diabetes, periodontitis and cardiovascular disease. Diabetes Metab Syndr. 13:1675–1678. 2019.PubMed/NCBI View Article : Google Scholar

9 

Trikka D and Vassilopoulos S: Periodontal regeneration with enamel matrix derivative in the management of generalized aggressive periodontitis: A case report with 11-year follow-up and literature review. J Int Soc Prev Community Dent. 9:13–20. 2019.PubMed/NCBI View Article : Google Scholar

10 

Bella P and Istvan G: The comprehensive periodontal, resorative end prosthodontic therapy of chronic periodontitis case presentation. Fogorv Sz. 109:125–135. 2016.PubMed/NCBI(In English, Hungarian).

11 

Ashouri Moghaddam A, Radafshar G, Jahandideh Y and Kakaei N: Clinical evaluation of effects of local application of aloe vera gel as an adjunct to scaling and root planning in patients with chronic periodontitis. J Dent (Shiraz). 18:165–172. 2017.PubMed/NCBI

12 

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.PubMed/NCBI View Article : Google Scholar

13 

Aydin S and Şahin F: Stem cells derived from dental tissues. Adv Exp Med Biol. 1144:123–132. 2019.PubMed/NCBI View Article : Google Scholar

14 

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.PubMed/NCBI View Article : Google Scholar

15 

Nagata M, Iwasaki K, Akazawa K, Komaki M, Yokoyama N, Izumi Y and Morita I: Conditioned medium from periodontal ligament stem cells enhances periodontal regeneration. Tissue Eng Part A. 23:367–377. 2017.PubMed/NCBI View Article : Google Scholar

16 

Diomede F, D'Aurora M, Gugliandolo A, Merciaro I, Ettorre V, Bramanti A, Piattelli A, Gatta V, Mazzon E, Fontana A and Trubiani O: A novel role in skeletal segment regeneration of extracellular vesicles released from periodontal-ligament stem cells. Int J Nanomed. 13:3805–3825. 2018.PubMed/NCBI View Article : Google Scholar

17 

Trubiani O, Marconi GD, Pierdomenico SD, Piattelli A, Diomede F and Pizzicannella J: Human oral stem cells, biomaterials and extracellular vesicles: A promising tool in bone tissue repair. Int J Mol Sci. 20(4987)2019.PubMed/NCBI View Article : Google Scholar

18 

Ramenzoni LL, Russo G, Moccia MD, Attin T and Schmidlin PR: Periodontal bacterial supernatants modify differentiation, migration and inflammatory cytokine expression in human periodontal ligament stem cells. PLoS One. 14(e0219181)2019.PubMed/NCBI View Article : Google Scholar

19 

Cheng M and Zhou Q: Targeting EZH2 ameliorates the LPS-inhibited PDLSC osteogenesis via Wnt/β-catenin pathway. Cells Tissues Organs. 209:227–235. 2020.PubMed/NCBI View Article : Google Scholar

20 

Wang W, Yuan C, Geng T, Liu Y, Zhu S, Zhang C, Liu Z and Wang P: Lipopolysaccharide inhibits osteogenic differentiation of periodontal ligament stem cells partially through toll-like receptor 4-mediated ephrinB2 downregulation. Clin Oral Investig. 24:3407–3416. 2020.PubMed/NCBI View Article : Google Scholar

21 

Fu Z, Wang X, Li B and Tang Y: Fraxinellone alleviates inflammation and promotes osteogenic differentiation in lipopolysaccharide-stimulated periodontal ligament stem cells by regulating the bone morphogenetic protein 2/Smad pathway. Arch Oral Biol. 121(104927)2021.PubMed/NCBI View Article : Google Scholar

22 

Honda S and Harigai M: The safety of baricitinib in patients with rheumatoid arthritis. Expert Opin Drug Saf. 19:545–551. 2020.PubMed/NCBI View Article : Google Scholar

23 

Ancuța C, Pomîrleanu C, Mihailov C, Chirieac R, Ancuța E, Iordache C, Bran C and Țănculescu O: Efficacy of baricitinib on periodontal inflammation in patients with rheumatoid arthritis. Joint Bone Spine. 87:235–239. 2020.PubMed/NCBI View Article : Google Scholar

24 

Murakami K, Kobayashi Y, Uehara S, Suzuki T, Koide M, Yamashita T, Nakamura M, Takahashi N, Kato H, Udagawa N and Nakamura Y: A Jak1/2 inhibitor, baricitinib, inhibits osteoclastogenesis by suppressing RANKL expression in osteoblasts in vitro. PLoS One. 12(e0181126)2017.PubMed/NCBI View Article : Google Scholar

25 

Adam S, Simon N, Steffen U, Andes FT, Scholtysek C, Müller DIH, Weidner D, Andreev D, Kleyer A, Culemann S, et al: JAK inhibition increases bone mass in steady-state conditions and ameliorates pathological bone loss by stimulating osteoblast function. Sci Transl Med. 12(eaay4447)2020.PubMed/NCBI View Article : Google Scholar

26 

Zheng XY, Mao CY, Qiao H, Zhang X, Yu L, Wang TY and Lu EY: Plumbagin suppresses chronic periodontitis in rats via down-regulation of TNF-α, IL-1β and IL-6 expression. Acta Pharmacol Sin. 38:1150–1160. 2017.PubMed/NCBI View Article : Google Scholar

27 

Shao Q, Liu S, Zou C and Ai Y: Effect of LSD1 on osteogenic differentiation of human periodontal ligament stem cells in periodontitis. Oral Dis: Nov 5, 2021 (Epub ahead of print).

28 

Zheng X, Zhu Y, Wang X, Hou Y and Fang Y: Silencing of ITGB6 inhibits the progression of cervical carcinoma via regulating JAK/STAT3 signaling pathway. Ann Transl Med. 9(803)2021.PubMed/NCBI View Article : Google Scholar

29 

Eruslanov E and Kusmartsev S: Identification of ROS using oxidized DCFDA and flow-cytometry. Methods Mol Biol. 594:57–72. 2010.PubMed/NCBI View Article : Google Scholar

30 

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.PubMed/NCBI View Article : Google Scholar

31 

Sanz M, Marco Del Castillo A, Jepsen S, Gonzalez-Juanatey JR, D'Aiuto F, Bouchard P, Chapple I, Dietrich T, Gotsman I, Graziani F, et al: Periodontitis and cardiovascular diseases: Consensus report. J Clin Periodontol. 47:268–288. 2020.PubMed/NCBI View Article : Google Scholar

32 

Lundmark A, Hu YOO, Huss M, Johannsen G, Andersson AF and Yucel-Lindberg T: Identification of salivary microbiota and its association with host inflammatory mediators in periodontitis. Front Cell Infect Microbiol. 9(216)2019.PubMed/NCBI View Article : Google Scholar

33 

Menicanin D, Mrozik KM, Wada N, Marino V, Shi S, Bartold PM and Gronthos S: Periodontal-ligament-derived stem cells exhibit the capacity for long-term survival, self-renewal, and regeneration of multiple tissue types in vivo. Stem Cells Dev. 23:1001–1011. 2014.PubMed/NCBI View Article : Google Scholar

34 

Li L, Liu W, Wang H, Yang Q, Zhang L, Jin F and Jin Y: Mutual inhibition between HDAC9 and miR-17 regulates osteogenesis of human periodontal ligament stem cells in inflammatory conditions. Cell Death Dis. 9(480)2018.PubMed/NCBI View Article : Google Scholar

35 

Shirasugi M, Nakagawa M, Nishioka K, Yamamoto T, Nakaya T and Kanamura N: Relationship between periodontal disease and butyric acid produced by periodontopathic bacteria. Inflamm Regen. 38(23)2018.PubMed/NCBI View Article : Google Scholar

36 

Hoare A, Soto C, Rojas-Celis V and Bravo D: Chronic inflammation as a link between periodontitis and carcinogenesis. Mediators Inflamm. 2019(1029857)2019.PubMed/NCBI View Article : Google Scholar

37 

Zhao B, Zhang W, Xiong Y, Zhang Y, Zhang D and Xu X: Effects of rutin on the oxidative stress, proliferation and osteogenic differentiation of periodontal ligament stem cells in LPS-induced inflammatory environment and the underlying mechanism. J Mol Histol. 51:161–171. 2020.PubMed/NCBI View Article : Google Scholar

38 

Duan Y, An W, Wu H and Wu Y: Salvianolic acid C attenuates LPS-induced inflammation and apoptosis in human periodontal ligament stem cells via toll-like receptors 4 (TLR4)/nuclear factor kappa B (NF-κB) pathway. Med Sci Monit. 25:9499–9508. 2019.PubMed/NCBI View Article : Google Scholar

39 

Smolen JS, Genovese MC, Takeuchi T, Hyslop DL, Macias WL, Rooney T, Chen L, Dickson CL, Riddle Camp J, Cardillo TE, et al: Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol. 46:7–18. 2019.PubMed/NCBI View Article : Google Scholar

40 

Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, et al: JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest. 128:3041–3052. 2018.PubMed/NCBI View Article : Google Scholar

41 

Bhaskarmurthy DH and Prince SE: Effect of baricitinib on TPA-induced psoriasis like skin inflammation. Life Sci. 279(119655)2021.PubMed/NCBI View Article : Google Scholar

42 

Douglas TEL, Vandrovcová M, Kročilová N, Keppler JK, Zárubová J, Skirtach AG and Bačáková L: Application of whey protein isolate in bone regeneration: Effects on growth and osteogenic differentiation of bone-forming cells. J Dairy Sci. 101:28–36. 2018.PubMed/NCBI View Article : Google Scholar

43 

Jiang B, Xu J, Zhou Y, Mao J, Guan G, Xu X and Mei L: Estrogen enhances osteogenic differentiation of human periodontal ligament stem cells by activating the Wnt/β-catenin signaling pathway. J Craniofac Surg. 31:583–587. 2020.PubMed/NCBI View Article : Google Scholar

44 

Yu M, Wang L, Ba P, Li L, Sun L, Duan X, Yang P, Yang C and Sun Q: Osteoblast progenitors enhance osteogenic differentiation of periodontal ligament stem cells. J Periodontol. 88:e159–e168. 2017.PubMed/NCBI View Article : Google Scholar

45 

Sun H, Feng K, Hu J, Soker S, Atala A and Ma PX: Osteogenic differentiation of human amniotic fluid-derived stem cells induced by bone morphogenetic protein-7 and enhanced by nanofibrous scaffolds. Biomaterials. 31:1133–1139. 2010.PubMed/NCBI View Article : Google Scholar

46 

Deng Y, Wu S, Zhou H, Bi X, Wang Y, Hu Y, Gu P and Fan X: Effects of a miR-31, Runx2, and Satb2 regulatory loop on the osteogenic differentiation of bone mesenchymal stem cells. Stem Cells Dev. 22:2278–2286. 2013.PubMed/NCBI View Article : Google Scholar

47 

Li J, Li L, Wang X and Xiao L: Porphyromonas gingivalis inhibition of MicroRNA-205-5p expression modulates proinflammatory cytokines in gingival epithelial cells. Biochem Genet. 58:566–579. 2020.PubMed/NCBI View Article : Google Scholar

48 

Wang Q, Li H, Xie H, Fu M, Guo B, Ding Y, Li W and Yu H: 25-Hydroxyvitamin D3 attenuates experimental periodontitis through downregulation of TLR4 and JAK1/STAT3 signaling in diabetic mice. J Steroid Biochem Mol Biol. 135:43–50. 2013.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

February-2023
Volume 25 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang P, Shi F and Zhang Y: Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling. Exp Ther Med 25: 74, 2023
APA
Yang, P., Shi, F., & Zhang, Y. (2023). Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling. Experimental and Therapeutic Medicine, 25, 74. https://doi.org/10.3892/etm.2022.11773
MLA
Yang, P., Shi, F., Zhang, Y."Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling". Experimental and Therapeutic Medicine 25.2 (2023): 74.
Chicago
Yang, P., Shi, F., Zhang, Y."Baricitinib alleviates lipopolysaccharide‑induced human periodontal ligament stem cell injury and promotes osteogenic differentiation by inhibiting JAK/STAT signaling". Experimental and Therapeutic Medicine 25, no. 2 (2023): 74. https://doi.org/10.3892/etm.2022.11773