Next Article in Journal
Evaluation of Physicochemical and Mechanical Properties of a Modified Adhesive System by Resveratrol Incorporation
Previous Article in Journal
Animal Experimental Study on Delayed Implantation in a Severely Atrophic Alveolar Ridge Reconstructed Using a 3D-Printed Bioactive Glass Scaffold: A Pilot Study
Previous Article in Special Issue
Anti-Aging Effects and Mechanisms of Cod Collagen Peptides (CCPs) in Caenorhabditis elegans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metformin-Enhanced Secretome from Periodontal Ligament Stem Cells Promotes Functional Recovery in an Inflamed Periodontal Model: In Vitro Study

1
Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup 56212, Jeollabuk-do, Republic of Korea
2
Department of Maxillofacial Biomedical Engineering, College of Dentistry, Kyung Hee University, 26 Kyunghee-daero, Dongdaemun-gu, Seoul 02447, Republic of Korea
*
Author to whom correspondence should be addressed.
J. Funct. Biomater. 2025, 16(5), 177; https://doi.org/10.3390/jfb16050177
Submission received: 24 March 2025 / Revised: 28 April 2025 / Accepted: 10 May 2025 / Published: 13 May 2025
(This article belongs to the Special Issue Natural Biomaterials for Biomedical Applications)

Abstract

:
Objective: Secretory factors, termed the secretome, in the conditioned medium (CM) from dental mesenchymal stem cells (MSCs) have shown anti-inflammatory, anti-apoptotic, and tissue regenerative potential. This cell-free product could be further developed by preconditioning cells with various biochemical agents, which lead to a change in secretome and CM profiles. Among the favorable candidates for CM production, metformin as an anti-diabetic medication is currently considered a potential agent for dental hard tissue and periodontal regeneration. Here, we aimed to assess the composition of CM from periodontal ligament stem cells (PDLSCs) grown in metformin-preconditioned media (Met-CM) compared to normal PDLSC-CM and assess the ability of Met-CM to recover the function of inflamed PDLSCs. Methods: Met-CM and normal CM were collected from PDLSCs grown with or without 50 µM metformin, respectively, under healthy culture conditions. Mass spectrometry and liquid chromatography–tandem mass spectrometry (LC–MS/MS) were performed to comparatively evaluate the proteomic profiles in PDLSC-CM versus Met-CM. We then treated the PDLSC cultures with lipopolysaccharide (LPS) from Porphyromonas gingivalis to induce inflammation and evaluated the osteogenic/cementogenic differentiation in the presence of Met-CM or normal PDLSC-CM by assessing alkaline phosphatase activity, intracellular calcium levels, and mRNA expression of osteogenic and cementogenic factors, including RUNX2, OCN, OSX, and CEMP-1. Subsequently, we performed RNA sequencing to identify transcriptomic changes in the treated cells. Results: We identified 202 differentially expressed proteins, 175 of which were significant, in Met-CM versus normal PDLSC-CM. Among the analyzed groups, the top three protein classes were protein-binding activity modulator, cytoskeletal protein, and extracellular matrix (ECM) protein. Treatment of PDLSCs with LPS significantly attenuated ALP activity, [Ca2+]i, and the mRNA expression levels of RUNX2, OCN, OSX, and CEMP-1, whereas treatment with Met-CM alone markedly enhanced PDLSC differentiation activity compared with the control. Moreover, osteogenic/cementogenic differentiation of the LPS-treated PDLSCs was recovered through incubation in Met-CM. Transcriptomic analysis identified 511 and 3591 differentially expressed genes in the control versus Met-CM and LPS versus LPS + Met-CM groups, respectively. The enrichment of biological processes includes positive regulation of DNA-templated transcription and skeletal system morphogenesis in the control versus Met-CM comparison, as well as positive regulation of transcription from the RNA polymerase II promoter and negative regulation of the apoptotic process in the LPS versus LPS + Met-CM comparison. Molecular function analysis demonstrated the enrichment of protein-binding terms among the DEGs from each comparison. Conclusions: Metformin preconditioning enhanced the recovery effect of PDLSC-CM on LPS-induced inflamed PDLSCs. These findings suggest that metformin preconditioning could represent a practical formula for PDLSC-secretome, which may contribute to the development of future cell-free periodontal regenerative strategies.

1. Introduction

It is widely recognized that dental stem cells such as dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), dental follicle cells (DFCs), and stem cells from human pulp of exfoliated deciduous teeth (SHED) and apical papilla (SCAPs) are a key component of regenerative dentistry, with the competence to release growth factors, cytokines, and various signaling molecules that are thought to promote dental tissue regeneration [1,2,3]. These stem cell-derived paracrine factors, referred to as the secretome, in conditioned medium (CM) include tissue-protective and regenerative molecules that can act as cell-free therapeutics [4]. A trend toward the use of stem cell CM in periodontal tissue regeneration research is also emerging, with results suggesting that indirect stem cell therapy shows effects consistent with those of stem cell transplantation [5,6]. Among the possible sources of dental stem cell CM, periodontal ligament stem cells (PDLSCs) are most effective at supplying a large number of secretory molecules for periodontal regeneration. It was reported that PDLSC-derived CM (PDLSC-CM) promotes regenerative and anti-inflammatory effects in various tissues, including periodontal, chondrogenic, and osteogenic systems [2,3,7].
Because CM mirrors the state of the cell culture liquid, CM composition is affected by various physical and chemical preconditioning within the cell culture microenvironment [8]. Recent experiments have demonstrated that CM produced from cell cultures treated with certain bioactive molecules could modulate cellular activities in various experimental systems. For example, CM from macrophages incubated with a combination of several growth factors improved the phagocytosis of myelin debris and increased the expression of nerve repair-related genes in olfactory ensheathing cells [9]. CM derived from astrocytes cultured with ciliary neurotrophic factor was also found to contain potent neuroactive factors that significantly influence neuronal calcium channel activity [10]. In addition, CM from hypoxic effect-preconditioned human umbilical cord-derived mesenchymal stem cells was shown to induce increased secretion of angiogenic and neuro-protective factors relative to normal CM [11]. Although, to date, few reports have described the preconditioning processes for producing modified CM, particularly from PDLSCs, strategies for manufacturing PDLSC-CM with enhanced paracrine activities are required for the therapeutic use of cell-free CM in the field of periodontal regenerative medicine.
In several ways, modifying CM by adding specific chemical compounds to the culture medium can enhance specific cellular processes and target certain pathways to create desired therapeutic effects. Among the promising chemical candidates for CM production, this study explores chemicals, specifically in targeted periodontal tissue repair and regeneration. Metformin (1,1-dimethylbiguanide hydrochloride) is an oral anti-diabetic agent that is widely used for medication of type 2 diabetes [12]. It is further noted that this agent can promote the osteogenic differentiation of stem cells and osteogenic progenitor cells [12,13,14]. Regarding metformin’s effects on PDLSC activity, several studies recently demonstrated that direct treatment of PDLSC cultures with metformin promoted osteogenic differentiation [12,15,16]. Results from in vivo experimental models further showed accelerated formation of new bone in the peri-implant regions in animals treated with metformin compared with their control group, suggesting that metformin can enhance implant osseointegration [17,18]. When periodontitis was induced in a rat model, metformin treatment has shown attenuated inflammation and alveolar bone loss [19]. Emerging studies on diabetic periodontitis indicated that metformin encouraged reducing inflammation and less bone loss in periodontitis [20,21]. A clinical study also presented that applying gelated metformin into the periodontal pockets of patients with chronic periodontitis and bone defects significantly improved their clinical and radiological parameters [22].
Based on these previous works, metformin mainly exhibits dual capacities, with anti-diabetic coupled with anti-inflammation and osteogenic activities at both the cellular and tissue levels. Thus, it is worth challenging its considerable potential as a chemical preconditioning option to modify the culture conditions of PDLSCs, which could lead to a more targeted and potent secretome for promoting periodontal regeneration. However, not only has preconditioning PDLSCs with metformin rarely been reported, but it also has not been fully understood how metformin modifies the composition and functionality of the secreted factors and signaling molecules released from PDLSCs.
The present study hypothesized whether preconditioning PDLSC cultures with metformin could modify the content of CM toward increased levels of periodontal protective and regenerative factors and thus improve the therapeutic potential of normal PDLSC-CM for periodontal health. To test this hypothesis, this study was designed to (1) compare the secretory protein contents in modified CM obtained from metformin-preconditioned PDLSC culture (Met-CM) versus normal PDLSC-CM and (2) assess the effects of Met-CM on the differentiation capacity and molecular dynamics of PDLSCs under inflammatory conditions using RNA sequencing analysis. Our findings indicate that metformin preconditioning improved the secretome contents from PDLSCs and their CM product, and Met-CM promoted functional recovery in inflamed PDLSCs.

2. Materials and Methods

2.1. Periodontal Ligament Stem Cell Culture

Human PDLSCs (CELPROGEN, Torrance, CA, USA) were purchased and cultured in Minimum Essential Medium α (α-MEM; Gibco-BRL, Waltham, MA, USA) containing 10% fetal bovine serum (FBS; Gibco-BRL, Waltham, MA, USA) as previously reported [23]. The PDLSCs at passages 4–6 were used for all experiments. For each experiment, the basic α-MEM culture media was supplemented with 5% FBS, 50-μg/mL ascorbic acid, 1 μM dexamethasone, and 3 mM β-glycerophosphate, corresponding to the formula of osteogenic medium. Culture media were changed every 2 days. For experiments in which the PDLSC cultures were treated with Met-CM or lipopolysaccharide (LPS), cells were organized into the following four treatment groups: osteogenic medium (Control), osteogenic medium + LPS (5 μg/mL), Met-CM, and LPS + Met-CM. To correspond to the formula of osteogenic medium, Met-CM was complemented by the osteoinductive substances mentioned above for the Met-CM and LPS + Met-CM groups. All reagents and laboratory expendables were obtained from the Sigma Chemical Company (St. Louis, MO, USA) and SPL Lifescience (Pocheon, Republic of Korea), respectively.

2.2. Preparation of CM

PDLSC-CM and Met-CM were collected from cells grown with or without metformin (50 µM) under healthy culture conditions. In brief, PDLSCs were cultured to 70% confluency and then washed with phosphate-buffered saline (PBS), after which they were cultured with or without metformin in serum-free α-MEM in the absence of FBS and antibiotics for 48 h. Culture supernatants were then harvested and centrifuged at 3000 rpm for 5 min to remove cell debris. This obtained medium was referred to as PDLSC-CM or Met-CM for all experiments.

2.3. Protein Digestion

Prior to digestion, proteins were processed using filter-aided sample preparation on a Microcon 30 kDa Centrifugal Filter device (Millipore, Billerica, MA, USA) and reduced with Tris(2-carboxyethyl)phosphine at 37 °C for 30 min [24]. Proteins were alkylated with iodoacetic acid at 25 °C for 1 h in the dark and washed with lysis buffer and ammonium bicarbonate (50 mM). This sample was then treated and broken down into peptides with trypsin at 37 °C for 18 h. The peptides were desalted using C18 spin columns (Harvard Apparatus, Holliston, MA, USA) and eluted with 80% acetonitrile in 0.1% formic acid.

2.4. Liquid Chromatography–Tandem Mass Spectrometry (LC–MS/MS) Analysis

LC–MS/MS analysis was performed as described in a previous study [24]. Briefly, the peptide fragments were solubilized in 0.1% formic acid and analyzed using a Q-Exactive Orbitrap hybrid mass spectrometer (Thermo Fisher Scientific, Waltham, MA, USA) with an Ultimate 3000 system (Thermo Fisher Scientific). Peptides were separated according to their hydrophobicity using a 2 cm × 75 μm ID trap column packed with 3 μm C18 resin and a 50 cm × 75 μm ID analytical column packed with 2 μm C18 resin. After separation, data-dependent acquisition was performed, and the top 10 precursor peaks were selected for fragmentation. Ions were scanned at a high resolution (70,000 in MS1 and 17,500 in MS2 at m/z 400), with an MS scan range of 400–2000 m/z at both the MS1 and MS2 levels. Precursor ions were fragmented with 27% normalized collisional energy. Dynamic exclusion was adjusted to 30 s.

2.5. Proteome Data Analysis

Proteome data were analyzed as described in our previous study [25]. The MS/MS raw files of each analysis were explored using Proteome Discoverer™ software (ver. 2.5) and the Homo sapiens database (Uniprot). A peptide-spectrum match (PSM) identification and SEQUEST HT process were provided as a database search engine. The search parameters were established as follows: 10 ppm of tolerances of precursor ion masses, 0.02 Da fragment ion mass, and a maximum of two missed cleavages with trypsin. The dynamic modification of the peptide sequence was as follows: static carbamidomethylation of cysteine (+57.012 Da), variable modifications of methionine oxidation (+15.995 Da), acetylation of the protein’s N-term (+42.011 Da), and carbamylation of the protein’s N-term (+43.0006 Da). Among the accumulated findings, data below 1% of the FDR were selected and filtered at least 6 more peptide lengths.

2.6. Validation of DEPs by ELISA

An ELISA was performed as a validation tool for the differentially expressed proteins (DEPs) using an ELISA kit (AssayGenie, Dublin, Ireland). The experimental procedure was performed by following the manufacturer’s instructions. The optical density (OD) was measured spectrophotometrically at a wavelength of 450 nm.

2.7. Alkaline Phosphatase Activity

To assess the levels of PDLSC osteogenic differentiation, the alkaline phosphatase (ALP) activity was measured as previously described [23]. The extracted total was added to 200 μL of p-nitrophenyl phosphate (pNPP) and incubated for 30 min at 37 °C. The reaction was then stopped with 3-M NaOH solution, and the optical density was determined using spectrophotometry at 405 nm. ALP activity was presented in terms of mM/100 μg of protein.

2.8. Intracellular Calcium Quantification Assay

The intracellular calcium levels were measured as described in our previous report [23]. Briefly, cells were cultured in each experimental condition for 14 days, and the intracellular calcium concentration was measured using a QuantiChrom™ calcium assay kit (BioAssay Systems, Hayward, CA, USA) according to the manufacturer’s instructions. The calcium content of each sample was calculated in terms of mg/100 mg of protein, from which the optical density (OD) was measured at 612 nm.

2.9. RNA Extraction and Real-Time Reverse Transcription (qRT)-PCR

To examine the mRNA expression of osteogenic and cementogenic factors, and to validate the RNA sequencing results, RNA extraction, cDNA synthesis, and qRT-PCR were performed as previously described [23] and according to the instructions provided with the QuantiTect SYBR Green PCR Kit (QIAGEN) using an iCycler iQ Multi-Color Real-Time Detection System (Bio-Rad). The thermal cycling conditions were set to 95 °C for 30 s, 95 °C for 5 s, 55 °C for 30 s, and 72 °C for 30 s for 30 cycles. The primers were as follows: 5′-CCCAGTATGAGAGTAGGTGTCC-3′ (sense) and 5′-GGGTAAGACTGGTCATAGGACC-3′ (antisense) for RUNX2; 5′-CGCTACCTGTATCAATGGCTGG-3′ (sense) and 5′-CTCCTGAAAGCCGATGTGGTCA-3′ (antisense) for OCN; 5′-TTCTGCGGCAAGAGGTTCACTC-3′ (sense) and 5′-GTGTTTGCTCAGGTGGTCGCTT-3′ (antisense) for OSX; 5′-CCATCCTATCTCTTTGGACCTGG-3′ (sense) and 5′-CCTTGCTTACAGGTGCTGTCCT-3′ (antisense) for CEMP-1; and 5′-GCTCTCCAGAACATCATCC-3′ (sense) and 5′-TGCTTCACCACCTTCTTG-3′ (antisense) for GAPDH; 5′-ATGGTCACCTGGTCACTCCAAC-3′ (sense) and 5′-GAGGCACAGAAGCTGCAAAAGG-3′ (antisense) for OPN3; 5′-TGTATCTGCTCTCGGACAAGGC-3′ (sense) and 5′-GCTGAGGAAACTGCATTGGAACC-3′ (antisense) for TRPM4; 5′-CACTACCATCTGAACTGTGGCTG-3′ (sense) and 5′-GCTTTCGTTCCAACAGCCAGTC-3′ (antisense) for TIMP4; and 5′-GGCACAATGTCTCCTCCAGAGA-3′ (sense) and 5′-CAGATGAAGCCTTGGTCAGTGC-3′ (antisense) for FOXP3.

2.10. Library Preparation and Sequencing

The total RNA was extracted, and the RNA concentration was assessed. The construction of a library was prepared using a QuantSeq 3′ mRNA-Seq Library Prep Kit FWD (Lexogen) according to the manufacturer’s instructions [26]. In brief, reverse transcription was conducted with the hybridized product of the prepared total RNA of each sample and an oligo-dT primer with an Illumina-compatible sequence at its 5′ end. The RNA template was then degraded, and the second strand was initiated to synthesize with a random primer containing an Illumina-compatible linker sequence at its 5′ end. The double-stranded library was purified and amplified to add the necessary adapter sequences for cluster generation. The completed library was then purified to remove contamination from the PCR components. Eventually, high-throughput sequencing was performed in the form of single-end 75 bp sequencing using NextSeq 500 (Illumina Inc., San Diego, CA, USA).

2.11. Genome Data Analysis

FastQC was used to evaluate the quality control of the raw sequencing data [27]. Adapter sequences and low-quality reads were removed by using bbduk 38.34 [28]. The clean sequencing reads were aligned to a known reference genome sequence using Bowtie2 alignment software, specifically version 2.3.5.1 [29]. The reads were then quantified using Bedtools v2.27.1 [30]. The TMM+CPM normalization method using EdgeR was applied to the read counts [31]. Data mining and graphic visualization were established using ExDEGA (Ebiogen Inc., Seoul, Republic of Korea). The DAVID (http://david.abcc.ncifcrf.gov/, accessed on 31 January 2023) program was used to determine the gene classification, ontology, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. STRING v3.8.2 was employed to predict protein–protein interactions (PPIs), with a PPI score >0.4 (p < 0.05) considered to be significant. The PPI networks were also visualized using Cytoscape v3.10.0 software (http://www.cytoscape.org, accessed on 2 November 2023).

2.12. Statistical Analysis

Data are shown as the mean ± SD (n ≥ 3), and they were evaluated using the Student’s t-test. One-way analysis of variance was performed for multiple comparisons using Duncan’s multiple range test, where p values < 0.05 were considered statistically significant. The figures shown are representative of the data.

3. Results

3.1. Comparison of Differentially Expressed Proteins (DEPs) in PDLSC-CM Versus Met-CM

To identify proteins differentially expressed in Met-CM compared with PDLSC-CM, we collected CM from cultures of human PDLSCs grown in normal or metformin-preconditioned serum-free culture media. LC–MS/MS proteomics was then performed, uncovering a total of 202 DEPs. We further analyzed the DEPs and identified 175 significant protein changes based on a fold-change cut-off of ≥2 and p < 0.05 (Supplementary Data S1). Hierarchical clustering was performed to group significant DEPs, and dynamic changes in the DEPs were assessed, revealing that 17 proteins were increased and 158 proteins were decreased in Met-CM relative to PDLSC-CM (Figure 1A). The enzyme-linked immunosorbent assay (ELISA) results were used to validate the DEPs. The ELISA results yielded consistency with the proteomics data for three proteins: fibronectin, keratin type II cytoskeletal 1 (KRT1), and caspase-14 (Supplementary Data S2A). Subsequently, we used PANTHER to predict the potential roles of these proteins, and 149 of the 175 candidate DEPs recognized by the software were categorized based on their known functions (Figure 1B). Among the analyzed groups, the top three protein classes in 17 categories were the protein-binding activity modulator (PC00095), cytoskeletal protein (PC00085), and extracellular matrix (ECM) protein (PC00102). These protein groups were also identified by PANTHER subgroup classification (Supplementary Data S3). In total, 88 of the 175 DEPs recognized by PANTHER were assigned to molecular pathways, including integrin signaling, cytoskeletal regulation by Rho GTPase, and the Wnt signaling pathway (Figure 2). These comparative proteomics results indicate that the cellular secretome in Met-CM comprised many categories of proteins with the potential to modulate the molecular function of PDLSCs.

3.2. Effect of Met-CM on the Differentiation Capability of PDLSCs in LPS-Induced Inflammatory Conditions

We next evaluated the effect of Met-CM on the osteogenic and cementogenic differentiation of PDLSCs in an in vitro inflamed microenvironment. To this end, the cells were treated with osteogenic medium (as a control), osteogenic medium + LPS, Met-CM, or LPS + Met-CM. We then measured the osteogenic differentiation and found it to be significantly decreased in cells treated with LPS relative to the control group, as determined by ALP activity (Figure 3A) and intracellular calcium levels ([Ca2+]i) (Figure 3B). In addition, the mRNA expression levels of osteogenic and cementogenic factors, such as RUNX2, OCN, OSX, and CEMP-1 (Figure 3C), were downregulated by LPS treatment. In contrast, treatment with Met-CM significantly increased the osteogenic and cementogenic activity of PDLSCs compared with the control group. In addition, treatment with Met-CM alleviated the downregulation of osteogenic and cementogenic differentiation observed in the inflamed PDLSCs treated with LPS. These results indicate that Met-CM contains a PDLSC secretome that can efficiently heighten the differentiation potential of PDLSCs and therefore potentially influence periodontal tissue repair and regeneration.

3.3. Identification of DEGs and Gene Ontology (GO) Enrichment

To further investigate the effect of Met-CM on the global expression of genes and molecular pathways in PDLSCs, the cells in the control, LPS-, Met-CM-, and LPS + Met-CM-treated groups were analyzed by RNA sequencing. We then performed comparative analyses of the DEGs (fold-change ≥2 and p < 0.05) in the control versus Met-CM and LPS versus LPS + Met-CM groups. From these two comparisons, we identified 511 (279 upregulated and 232 downregulated) and 3591 (1745 upregulated and 1846 downregulated) DEGs, respectively (Figure 4A,B, Supplementary Data S4). The genomic profile was then validated through qRT-PCR, confirming the results obtained from RNA sequencing (Supplementary Data S2B). We then classified the DEGs into specific functional categories, including cell differentiation, cell cycle, angiogenesis, ECM, secretion, bone development, bone morphogenesis, cellular response to stress, immune response, and inflammatory response in the different comparisons (Figure 4C). The DEGs identified from each comparison were also analyzed by GO classification to identify enriched biological processes and molecular functions (Figure 5, Supplementary Data S5 and S6). The DEGs were specifically enriched in biological processes, including positive regulation of DNA-templated transcription and skeletal system morphogenesis in the control versus Met-CM comparison and positive regulation of transcription from the RNA polymerase II promoter and negative regulation of the apoptotic process in the LPS versus LPS + Met-CM comparison. Molecular function analysis revealed the enrichment of protein-binding terms among the DEGs from each comparison. These comparative analyses of the DEGs identified above, combined with functional annotation, reveal the molecular biomarker profiles produced in response to Met-CM treatment in healthy or inflamed PDLSC cultures.

3.4. KEGG Enrichment Pathway Analysis

The signaling pathways enriched among the DEGs identified from the two comparisons were further analyzed using the DAVID program; the top 10 significantly enriched pathways from each comparison are shown in Table 1.
Among the KEGG pathways identified, intriguing candidates with a possible role in bone and tooth mineralization and repair [32,33] included inositol phosphate metabolism for DEGs from the control versus Met-CM comparison (Figure 6) and protein processing in the endoplasmic reticulum (ER) for DEGs from the LPS versus LPS + Met-CM comparison (Figure 7).

3.5. Identification of PPI Networks

Lastly, we uploaded the DEGs from the two comparisons to the STRING online database and constructed putative PPI networks for each group. We detected 491 nodes (representing proteins) and 857 edges (representing interactions) in the control versus Met-CM network and 3427 nodes and 58,791 edges in the LPS versus LPS + Met-CM network. The top five hub proteins in each comparison are listed in Table 2.
From these PPI network data, we identified and visualized the meaningful clusters in each comparison, including mitogen-activated protein kinase 3 (MAPK3) and cyclin A2 (CCNA2) in the control versus Met-CM group and AKT1 substrate 1 (AKT1) and catenin beta 1 (CTNNB1) in the LPS versus LPS + Met-CM group using Cytoscape software (Figure 8).

4. Discussion

This study has shown how metformin modifies and potentiates the secretome and CM of PDLSCs and Met-CM encourages functional recovery of inflamed PDLSCs. Regarding the potent secretome contents to advance periodontal regeneration, comparative proteomics analysis revealed a high proportion of cytoskeletal and ECM proteins in Met-CM compared with the normal PDLSC-CM. These findings may be related to the signaling pathways identified, including integrin signaling, cytoskeletal regulation by Rho GTPase, and the Wnt signaling pathway. It is well known that the cytoskeleton, including microtubules, actin fibers, and intermediate filaments, interacts with proteins of the ECM in cell-to-matrix and cell-to-cell adhesions, as well as in various cellular processes, such as proliferation, migration, differentiation, and apoptosis [34]. In periodontal tissues, resident stem and progenitor cells commonly induce complex signaling pathways that regulate cellular components ranging from the ECM to the cytoskeleton and the nucleus, regarded as mechanoresponsive cellular processes [35,36,37]. Furthermore, the interplay among periodontal ECM components and cytoskeletal structures contributes to periodontal health and tissue regeneration [38,39]. Thus, our results suggest that the Met-CM containing concentrated cytoskeletal and ECM contents could serve as a possible therapeutic agent for periodontal tissue diseases and oral regenerative medicine.
This study further demonstrated the ability of Met-CM to promote the recovery of activities in inflamed PDLSCs. Using genomic profiling, we also analyzed the biological processes and dynamic molecular pathways enriched among DEGs identified from two comparative groups—PDLSCs treated with the control versus Met-CM and LPS versus LPS + Met-CM—to clearly define the effects of Met-CM in healthy or inflamed PDLSCs. KEGG pathway analysis revealed that the DEGs in the PDLSCs treated with Met-CM versus the control group were enriched for inositol phosphate metabolism. The inositol-derived metabolites, known as inositol polyphosphates, have a broad range of physiological functions and cellular activities, including DNA damage repair, energy production, and calcium homeostasis [40,41,42]. Numerous prior studies have suggested that metformin acts through the inositol phosphate signaling network and the phospholipase C/inositol 1,4,5-trisphosphate (IP3)/Ca2+ signaling pathway [43,44], which is also known to be a critical cellular cascade for bone and tooth mineralization and formation [45,46].
Our KEGG analysis also identified the enrichment of protein processing in the ER in LPS-stimulated PDLSCs treated with Met-CM. Protein processing in the ER ensures correct protein conformation and quality for maintaining protein homeostasis [47]. Protein homeostasis can be disrupted in cells responding to a variety of pathological and physiological conditions that induce ER stress. Consequently, cells will attempt to restore homeostasis through the downstream unfolded protein response (UPR) pathway, which comprises protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6) [48]. Previous studies have shown that the IRE1α-X-box binding protein (XBP1), PERK-eukaryotic initiation factor 2α (eIF2α), and ATF6 signaling pathways induce the UPR and restore ER stability [49,50]. However, the role of ER stress signaling pathways in the periodontal healing process is not clearly defined. In contrast, most studies have reported that ER stress signaling pathways are involved in cell apoptosis and death and act as key players in various human diseases [51,52,53]. One recent study reported that ER stress stimulates osteogenic responses and alveolar bone formation in tooth extraction sockets [33]. Moreover, another interesting investigation found that ER stress-activated PERK-eIF2α-ATF4 pathways promote osteogenic differentiation of human periodontal ligament cells under conditions of mechanical stress [46]. Our results here, together with those from previous studies, may indicate that the ER system is dynamically regulated and influenced in unconventional ways under specific pathological conditions, resulting in the healing or degeneration of periodontal tissue. However, further research is required to elucidate the possible association between the ER organelle and periodontal biological signaling.
The results from our PPI network analysis further identified MAPK3 and cyclin A2 in the control versus Met-CM-treated PDLSCs. These highly interacting and abundant proteins are widely known as signaling molecules that regulate various cellular processes, such as proliferation, differentiation, and cell cycle progression, in response to various extracellular signals [54,55,56]. In the LPS versus LPS + Met-CM groups, the notable hub proteins identified were AKT1 and CTNNB1, which are regarded as positive regulators of osteogenic differentiation of stem cells and bone mineralization and formation, as well as embryonic development and organogenesis and adult tissue homeostasis [57,58,59,60]. These clustering analyses could indicate that Met-CM treatment in healthy or inflamed PDLSC cultures promotes enhanced tissue growth and regeneration dynamics, suggesting potential therapeutic roles and biomarkers for periodontal diseases and defects.
Although this study suggests novel Met-CM efficacy in periodontal regeneration, it is still limited within the scope of an in vitro periodontal model. Given that periodontal biological and pathological events are regulated by the complex interactions between cells and their resident tissue environments, further research would be required to validate the impact of Met-CM on periodontal tissues with preclinical or clinical periodontal disease. While these proteomics and genomics studies confirmed various proteins, genes, and signaling pathways which could be involved in restoring impaired functions of PDLSCs, further examination with the interconnecting mechanisms between individual molecules could strengthen the present findings. To obtain more abundant protein or gene outcomes from the proteomics and genomics techniques, a valid strategy to produce concentrated formulations of CM can be employed in further experiments.
In conclusion, these findings suggest that Met-CM could represent a beneficial formula with regenerative potential for periodontal tissue and highlight encouraging functional recovery of inflamed PDLSCs. The modified and enriched PDLSC-CM as a cell-free product can be used in various periodontal therapeutic applications, including tissue engineering, cell-based therapies, and regenerative dentistry after the establishment of quality control criteria and manufacturing pharmaceutical standardization for the intended composition and concentration of CM.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jfb16050177/s1, Supplementary Data S1: LC-MS/MS data; Supplementary Data S2A: ELISA results to validate the DEPs; Supplementary Data S2B: qRT-PCR data to validate the DEGs; Supplementary Data S3: PANTHER subgroup classification; Supplementary Data S4; RNA sequencing-DEG data; Supplementary Data S5: GO classification to identify enriched biological processes; Supplementary Data S6: GO classification to identify enriched molecular functions.

Author Contributions

Conceptualization and design, J.S.H. and H.N.S.; methodology and investigation, J.Y.J.; data Curation, J.S.H. and H.N.S.; writing—original draft preparation, J.S.H. and H.N.S.; visualization, J.Y.J. and H.N.S.; supervision, J.S.H.; project administration, J.S.H. and H.N.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT; No. 2021R1A2C1014077) and the Korea Institute of Toxicology (KK-2505).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article and Supplementary Materials. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhai, Q.; Dong, Z.; Wang, W.; Li, B.; Jin, Y. Dental stem cell and dental tissue regeneration. Front. Med. 2019, 13, 152. [Google Scholar] [CrossRef] [PubMed]
  2. Nagata, M.; Iwasaki, K.; Akazawa, K.; Komaki, M.; Yokoyama, N.; Izumi, Y.; Morita, I. Conditioned Medium from Periodontal Ligament Stem Cells Enhances Periodontal Regeneration. Tissue Eng. Part. A 2017, 23, 367. [Google Scholar] [CrossRef] [PubMed]
  3. Qiu, J.; Wang, X.; Zhou, H.; Zhang, C.; Wang, Y.; Huang, J.; Liu, M.; Yang, P.; Song, A. Enhancement of periodontal tissue regeneration by conditioned media from gingiva-derived or periodontal ligament-derived mesenchymal stem cells: A comparative study in rats. Stem Cell Res. Ther. 2020, 11, 42. [Google Scholar] [CrossRef]
  4. Kumar, P.; Kandoi, S.; Misra, R.; Vijayalakshmi, S.; Rajagopal, K.; Verma, R.S. The mesenchymal stem cell secretome: A new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev. 2019, 46, 1. [Google Scholar]
  5. Gugliandolo, A.; Diomede, F.; Pizzicannella, J.; Chiricosta, L.; Trubiani, O.; Mazzon, E. Potential Anti-Inflammatory Effects of a New Lyophilized Formulation of the Conditioned Medium Derived from Periodontal Ligament Stem Cells. Biomedicines 2022, 10, 683. [Google Scholar] [CrossRef]
  6. Novello, S.; Tricot-Doleux, S.; Novella, A.; Pellen-Mussi, P.; Jeanne, S. Influence of Periodontal Ligament Stem Cell-Derived Conditioned Medium on Osteoblasts. Pharmaceutics 2022, 14, 729. [Google Scholar] [CrossRef]
  7. Rajan, T.S.; Giacoppo, S.; Trubiani, O.; Diomede, F.; Piattelli, A.; Bramanti, P.; Mazzon, E. Conditioned medium of periodontal ligament mesenchymal stem cells exert anti-inflammatory effects in lipopolysaccharide-activated mouse motoneurons. Exp. Cell Res. 2016, 349, 152. [Google Scholar] [CrossRef]
  8. Rosochowicz, M.A.; Lach, M.S.; Richter, M.; Suchorska, W.M.; Trzeciak, T. Conditioned Medium—Is it an Undervalued Lab Waste with the Potential for Osteoarthritis Management? Stem Cell Rev. Rep. 2023, 19, 1185. [Google Scholar] [CrossRef]
  9. Basu, S.; Choudhury, I.N.; Lee, J.Y.P.; Chacko, A.; Ekberg, J.A.K.; St John, J.A. Macrophages Treated with VEGF and PDGF Exert Paracrine Effects on Olfactory Ensheathing Cell Function. Cells 2022, 11, 2408. [Google Scholar] [CrossRef]
  10. Sun, M.; Liu, H.; Xu, H.; Wang, H.; Wang, X. CNTF-Treated Astrocyte Conditioned Medium Enhances Large-Conductance Calcium-Activated Potassium Channel Activity in Rat Cortical Neurons. Neurochem. Res. 2016, 41, 1982. [Google Scholar] [CrossRef]
  11. Ozkan, S.; Isildar, B.; Ercin, M.; Gezginci-Oktayoglu, S.; Konukoglu, D.; Neşetoğlu, N.; Oncul, M.; Koyuturk, M. Therapeutic potential of conditioned medium obtained from deferoxamine preconditioned umbilical cord mesenchymal stem cells on diabetic nephropathy model. Stem Cell Res. Ther. 2022, 13, 438. [Google Scholar] [CrossRef]
  12. Yang, Z.; Gao, X.; Zhou, M.; Kuang, Y.; Xiang, M.; Li, J.; Song, J. Effect of metformin on human periodontal ligament stem cells cultured with polydopamine-templated hydroxyapatite. Eur. J. Oral Sci. 2019, 127, 210. [Google Scholar] [CrossRef]
  13. Yang, K.; Cao, F.; Qiu, S.; Jiang, W.; Tao, L.; Zhu, Y. Metformin Promotes Differentiation and Attenuates H2O2-Induced Oxidative Damage of Osteoblasts via the PI3K/AKT/Nrf2/HO-1 Pathway. Front. Pharmacol. 2022, 13, 829830. [Google Scholar] [CrossRef]
  14. Smieszek, A.; Tomaszewski, K.A.; Kornicka, K.; Marycz, K. Metformin Promotes Osteogenic Differentiation of Adipose-Derived Stromal Cells and Exerts Pro-Osteogenic Effect Stimulating Bone Regeneration. J. Clin. Med. 2018, 7, 482. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, R.; Liang, Q.; Kang, W.; Ge, S. Metformin facilitates the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells in vitro. Cell Biol. Int. 2020, 44, 70. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, Y.L.; Liu, F.; Li, Z.B.; He, X.T.; Li, X.; Wu, R.X.; Sun, H.H.; Ge, S.H.; Chen, F.M.; An, Y. Metformin combats high glucose-induced damage to the osteogenic differentiation of human periodontal ligament stem cells via inhibition of the NPR3-mediated MAPK pathway. Stem Cell Res. Ther. 2022, 13, 305. [Google Scholar] [CrossRef] [PubMed]
  17. Yildirim, T.T.; Dundar, S.; Bozoglan, A.; Karaman, T.; Kahraman, O.E.; Ozcan, E.C. The effects of metformin on the bone filling ration around of TiAl6Va4 implants in non diabetic rats. J. Oral Biol. Craniofac. Res. 2020, 10, 474. [Google Scholar] [CrossRef]
  18. Lin, J.; Xu, R.; Shen, X.; Jiang, H.; Du, S. Metformin promotes the osseointegration of titanium implants under osteoporotic conditions by regulating BMSCs autophagy, and osteogenic differentiation. Biochem. Biophys. Res. Commun. 2020, 531, 228. [Google Scholar] [CrossRef]
  19. Araújo, A.A.; Pereira, A.S.B.F.; Medeiros, C.A.C.X.; Brito, G.A.C.; Leitão, R.F.C.; Araújo, L.S.; Guedes, P.M.M.; Hiyari, S.; Pirih, F.Q.; Araújo Júnior, R.F. Effects of metformin on inflammation, oxidative stress, and bone loss in a rat model of periodontitis. PLoS ONE 2017, 12, e0183506. [Google Scholar] [CrossRef]
  20. Pereira, A.S.B.F.; Brito, G.A.C.; Lima, M.L.S.; Silva Júnior, A.A.D.; Silva, E.D.S.; de Rezende, A.A.; Bortolin, R.H.; Galvan, M.; Pirih, F.Q.; Araújo Júnior, R.F.; et al. Metformin Hydrochloride-Loaded PLGA Nanoparticle in Periodontal Disease Experimental Model Using Diabetic Rats. Int. J. Mol. Sci. 2018, 19, 3488. [Google Scholar] [CrossRef]
  21. Zhou, X.; Zhang, P.; Wang, Q.; Ji, N.; Xia, S.; Ding, Y.; Wang, Q. Metformin ameliorates experimental diabetic periodontitis independently of mammalian target of rapamycin (mTOR) inhibition by reducing NIMA-related kinase 7 (Nek7) expression. J. Periodontol. 2019, 90, 1032. [Google Scholar] [CrossRef] [PubMed]
  22. Pradeep, A.R.; Patnaik, K.; Nagpal, K.; Karvekar, S.; Ramamurthy, B.L.; Naik, S.B. Efficacy of locally-delivered 1% metformin gel in the treatment of intrabony defects in patients with chronic periodontitis: A randomized, controlled clinical trial. J. Investig. Clin. Dent. 2016, 7, 239. [Google Scholar] [CrossRef]
  23. Shim, N.Y.; Ryu, J.I.; Heo, J.S. Osteoinductive function of fucoidan on periodontal ligament stem cells: Role of PI3K/Akt and Wnt/β-catenin signaling pathways. Oral Dis. 2022, 28, 1628–1639. [Google Scholar] [CrossRef] [PubMed]
  24. Nam, O.; Park, J.M.; Lee, H.; Jin, E. De novo transcriptome profile of coccolithophorid alga Emiliania huxleyi CCMP371 at different calcium concentrations with proteome analysis. PLoS ONE 2019, 14, e0221938. [Google Scholar] [CrossRef] [PubMed]
  25. Suh, H.N.; Ji, J.Y.; Heo, J.S. Translating proteome and transcriptome dynamics of periodontal ligament stem cell-derived secretome/conditioned medium in an in vitro model of periodontitis. BMC Oral Health 2024, 24, 390. [Google Scholar] [CrossRef]
  26. Kwack, K.H.; Ji, J.Y.; Park, B.; Heo, J.S. Fucoidan (Undaria pinnatifida)/Polydopamine Composite-Modified Surface Promotes Osteogenic Potential of Periodontal Ligament Stem Cells. Mar. Drugs 2022, 20, 181. [Google Scholar] [CrossRef]
  27. Li, K.; Zhang, S.; Gu, Y.; Wang, J.; Yang, Y.; Mao, W. Transcriptomic data of BT549 triple negative breast cancer cells treated with 20 M NU7441, a DNA-dependent kinase inhibitor. Data Brief 2024, 53, 110183. [Google Scholar] [CrossRef]
  28. Križanovic, K.; Echchiki, A.; Roux, J.; Šikic, M. Evaluation of tools for long read RNA-seq splice-aware alignment. Bioinformatics 2018, 34, 748. [Google Scholar] [CrossRef]
  29. Shinjo, K.; Umehara, T.; Niwa, H.; Sato, S.; Katsushima, K.; Sato, S.; Wang, X.; Murofushi, Y.; Suzuki, M.M.; Koyama, H.; et al. Novel pharmacologic inhibition of lysine-specific demethylase 1 as a potential therapeutic for glioblastoma. Cancer Gene Ther. 2024, 31, 1884. [Google Scholar] [CrossRef]
  30. Kiang, A.L.; Loo, S.S.; Mat-Isa, M.N.; Ng, C.L.; Blake, D.P.; Wan, K.L. Insights into genomic sequence diversity of the SAG surface antigen superfamily in geographically diverse Eimeria tenella isolates. Sci. Rep. 2024, 14, 26251. [Google Scholar] [CrossRef]
  31. Hejblum, B.P.; Ba, K.; Thiébaut, R.; Agniel, D. Neglecting the impact of normalization in semi-synthetic RNA-seq data simulations generates artificial false positives. Genome Biol. 2024, 25, 281. [Google Scholar] [CrossRef]
  32. Yang, Z.L.; Chen, J.N.; Lu, Y.Y.; Lu, M.; Wan, Q.L.; Wu, G.S.; Luo, H.R. Inositol polyphosphate multikinase IPMK-1 regulates development through IP3/calcium signaling in Caenorhabditis elegans. Cell Calcium. 2021, 93, 102327. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, Y.; Guo, Y.; Li, J.; Chen, Y.Y.; Liu, Q.; Tan, L.; Gao, Z.R.; Zhang, S.H.; Zhou, Y.H.; Feng, Y.Z. Endoplasmic reticulum stress remodels alveolar bone formation after tooth extraction. J. Cell Mol. Med. 2020, 24, 12411. [Google Scholar] [CrossRef] [PubMed]
  34. Tsunoyama, T.A.; Watanabe, Y.; Goto, J.; Naito, K.; Kasai, R.S.; Suzuki, K.G.N.; Fujiwara, T.K.; Kusumi, A. Super-long single-molecule tracking reveals dynamic-anchorage-induced integrin function. Nat. Chem. Biol. 2018, 14, 497. [Google Scholar] [CrossRef] [PubMed]
  35. Chukkapalli, S.S.; Lele, T.P. Periodontal cell mechanotransduction. Open Biol. 2018, 8, 180053. [Google Scholar] [CrossRef]
  36. Kechagia, J.Z.; Ivaska, J.; Roca-Cusachs, P. Integrins as biomechanical sensors of the microenvironment. Nat. Rev. Mol. Cell Biol. 2019, 20, 457. [Google Scholar] [CrossRef]
  37. Case, L.B.; Waterman, C.M. Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nat. Cell Biol. 2015, 17, 955. [Google Scholar] [CrossRef]
  38. Ugawa, Y.; Yamamoto, T.; Kawamura, M.; Yamashiro, K.; Shimoe, M.; Tomikawa, K.; Hongo, S.; Maeda, H.; Takashiba, S. Rho-kinase regulates extracellular matrix-mediated osteogenic differentiation of periodontal ligament cells. Cell Biol. Int. 2017, 41, 651. [Google Scholar] [CrossRef]
  39. Li, J.; Li, H.; Tian, Y.; Yang, Y.; Chen, G.; Guo, W.; Tian, W. Cytoskeletal binding proteins distinguish cultured dental follicle cells and periodontal ligament cells. Exp. Cell Res. 2016, 345, 6. [Google Scholar] [CrossRef] [PubMed]
  40. Yu, J.; Leibiger, B.; Yang, S.N.; Shears, S.B.; Leibiger, I.B.; Berggren, P.O.; Barker, C.J. Multiple Inositol Polyphosphate Phosphatase Compartmentalization Separates Inositol Phosphate Metabolism from Inositol Lipid Signaling. Biomolecules 2023, 13, 885. [Google Scholar] [CrossRef]
  41. Blind, R.D. Structural analyses of inositol phosphate second messengers bound to signaling effector proteins. Adv. Biol. Regul. 2020, 75, 100667. [Google Scholar] [CrossRef] [PubMed]
  42. Tu-Sekine, B.; Kim, S.F. The Inositol Phosphate System-A Coordinator of Metabolic Adaptability. Int. J. Mol. Sci. 2022, 23, 6747. [Google Scholar] [CrossRef]
  43. Shen, X.; Fan, B.; Hu, X.; Luo, L.; Yan, Y.; Yang, L. Metformin Reduces Lipotoxicity-Induced Meta-Inflammation in beta-Cells through the Activation of GPR40-PLC-IP3 Pathway. J. Diabetes Res. 2019, 2019, 7602427. [Google Scholar] [CrossRef] [PubMed]
  44. Jankeviciute, S.; Svirskiene, N.; Svirskis, G.; Borutaite, V. Effects of Metformin on Spontaneous Ca2+ Signals in Cultured Microglia Cells under Normoxic and Hypoxic Conditions. Int. J. Mol. Sci. 2021, 22, 9493. [Google Scholar] [CrossRef] [PubMed]
  45. Gao, X.; Di, X.; Li, J.; Kang, Y.; Xie, W.; Sun, L.; Zhang, J. Extracellular Calcium-Induced Calcium Transient Regulating the Proliferation of Osteoblasts through Glycolysis Metabolism Pathways. Int. J. Mol. Sci. 2023, 24, 4991. [Google Scholar] [CrossRef]
  46. Luo, B.; Luo, Y.; He, L.; Cao, Y.; Jiang, Q. Residual periodontal ligament in the extraction socket promotes the dentin regeneration potential of DPSCs in the rabbit jaw. Stem Cell Res. Ther. 2023, 14, 47. [Google Scholar] [CrossRef]
  47. Yu, Q.; Xiong, Y.; Gao, H.; Liu, J.; Chen, Z.; Wang, Q.; Wen, D. Comparative proteomics analysis of Spodoptera frugiperda cells during Autographa californica multiple nucleopolyhedrovirus infection. Virol. J. 2015, 12, 115. [Google Scholar] [CrossRef]
  48. Yang, S.Y.; Wei, F.L.; Hu, L.H.; Wang, C.L. PERK-eIF2α-ATF4 pathway mediated by endoplasmic reticulum stress response is involved in osteodifferentiation of human periodontal ligament cells under cyclic mechanical force. Cell Signal 2016, 28, 880. [Google Scholar] [CrossRef]
  49. Urra, H.; Dufey, E.; Avril, T.; Chevet, E.; Hetz, C. Endoplasmic reticulum stress and the hallmarks of cancer, trends. Cancer 2016, 2, 252. [Google Scholar]
  50. Lebeaupin, C.; Vallee, D.; Hazari, Y.; Hetz, C.; Chevet, E.; Bailly-Maitre, B. Endoplasmic reticulum stress signalling and the pathogenesis of nonalcoholic fatty liver disease. J. Hepatol. 2018, 69, 927. [Google Scholar] [CrossRef]
  51. Li, X.; Wang, Y.; Wang, H.; Huang, C.; Huang, Y.; Li, J. Endoplasmic reticulum stress is the crossroads of autophagy, inflammation, and apoptosis signaling pathways and participates in liver fibrosis. Inflamm. Res. 2015, 64, 1. [Google Scholar] [CrossRef] [PubMed]
  52. Miyazaki-Anzai, S.; Masuda, M.; Demos-Davies, K.M.; Keenan, A.L.; Saunders, S.J.; Masuda, R.; Jablonski, K.; Cavasin, M.A.; Kendrick, J.; Chonchol, M.; et al. Endoplasmic reticulum stress effector CCAAT/enhancer-binding protein homologous protein (CHOP) regulates chronic kidney disease-induced vascular calcification. J. Am. Heart Assoc. 2014, 3, e000949. [Google Scholar] [CrossRef] [PubMed]
  53. Sozen, E.; Karademir, B.; Ozer, N.K. Basic mechanisms in endoplasmic reticulum stress and relation to cardiovascular diseases. Free Radic. Biol. Med. 2015, 78, 30. [Google Scholar] [CrossRef] [PubMed]
  54. Ba, P.; Duan, X.; Fu, G.; Lv, S.; Yang, P.; Sun, Q. Differential effects of p38 and Erk1/2 on the chondrogenic and osteogenic differentiation of dental pulp stem cells. Mol. Med. Rep. 2017, 16, 63. [Google Scholar] [CrossRef]
  55. Wei, K.; Xie, Y.; Chen, T.; Fu, B.; Cui, S.; Wang, Y.; Cai, G.; Chen, X. ERK1/2 signaling mediated naringin-induced osteogenic differentiation of immortalized human periodontal ligament stem cells. Biochem. Biophys. Res. Commun. 2017, 489, 319. [Google Scholar] [CrossRef]
  56. Ye, J.; Ai, W.; Zhang, F.; Zhu, X.; Shu, G.; Wang, L.; Gao, P.; Xi, Q.; Zhang, Y.; Jiang, Q.; et al. Enhanced Proliferation of Porcine Bone Marrow Mesenchymal Stem Cells Induced by Extracellular Calcium is Associated with the Activation of the Calcium-Sensing Receptor and ERK Signaling Pathway. Stem Cells Int. 2016, 2016, 6570671. [Google Scholar] [CrossRef]
  57. Zhong, Y.T.; Liao, H.B.; Ye, Z.Q.; Jiang, H.S.; Li, J.X.; Ke, L.M.; Hua, J.Y.; Wei, B.; Wu, X.; Cui, L. Eurycomanone stimulates bone mineralization in zebrafish larvae and promotes osteogenic differentiation of mesenchymal stem cells by upregulating AKT/GSK-3beta/beta-catenin signaling. J. Orthop. Transl. 2023, 40, 132. [Google Scholar]
  58. Yang, J.; Zhang, L.; Ding, Q.; Zhang, S.; Sun, S.; Liu, W.; Liu, J.; Han, X.; Ding, C. Flavonoid-Loaded Biomaterials in Bone Defect Repair. Molecules 2023, 28, 6888. [Google Scholar] [CrossRef]
  59. Hu, L.; Chen, W.; Qian, A.; Li, Y.P. Wnt/beta-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res. 2024, 12, 39. [Google Scholar] [CrossRef]
  60. Steinhart, Z.; Angers, S. Wnt signaling in development and tissue homeostasis. Development 2018, 145, dev146589. [Google Scholar] [CrossRef]
Figure 1. Comparative proteomic analysis of proteins in condition medium (CM) from periodontal ligament stem cells (PDLSCs; PDLSC-CM) vs. those in CM from metformin-treated PDLSCs (Met-CM). (A) Hierarchical clustering of significantly differentially expressed proteins (fold-change ≥2 and p < 0.05). Colors depict relative expression levels of proteins, with red and blue indicating up- and downregulation, respectively. (B) Classification analysis of candidate proteins, categorized using the PANTHER program. The x axis shows the protein classification illustrated by different colors, and the number of proteins within each category is plotted on the y axis.
Figure 1. Comparative proteomic analysis of proteins in condition medium (CM) from periodontal ligament stem cells (PDLSCs; PDLSC-CM) vs. those in CM from metformin-treated PDLSCs (Met-CM). (A) Hierarchical clustering of significantly differentially expressed proteins (fold-change ≥2 and p < 0.05). Colors depict relative expression levels of proteins, with red and blue indicating up- and downregulation, respectively. (B) Classification analysis of candidate proteins, categorized using the PANTHER program. The x axis shows the protein classification illustrated by different colors, and the number of proteins within each category is plotted on the y axis.
Jfb 16 00177 g001
Figure 2. Signaling pathway enrichment analysis for 88 of the 175 differentially expressed proteins (DEPs) using the PANTHER program. (A) Distribution of proteins and (B) the list of pathways identified by PANTHER.
Figure 2. Signaling pathway enrichment analysis for 88 of the 175 differentially expressed proteins (DEPs) using the PANTHER program. (A) Distribution of proteins and (B) the list of pathways identified by PANTHER.
Jfb 16 00177 g002
Figure 3. Effects of lipopolysaccharide (LPS) and metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) on the osteogenic differentiation of PDLSCs. (A) Alkaline phosphatase (ALP) activity was measured after cells were cultured with LPS, Met-CM, or LPS + Met-CM for 7 days, and (B) [Ca2+]i was also assessed in same culture conditions for 14 days, as described under Materials and Methods. (C) The mRNA expression levels of RUNX2, OCN, OSX, and CEMP-1 were assessed by real-time reverse transcription (qRT)-PCR after 7 days of osteogenic induction. The values are shown as the mean ± standard deviation (n = 5). * p < 0.05 and ** p < 0.001 vs. the control value. # p < 0.05 and ## p < 0.001 vs. the Met-CM value at each time point.
Figure 3. Effects of lipopolysaccharide (LPS) and metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) on the osteogenic differentiation of PDLSCs. (A) Alkaline phosphatase (ALP) activity was measured after cells were cultured with LPS, Met-CM, or LPS + Met-CM for 7 days, and (B) [Ca2+]i was also assessed in same culture conditions for 14 days, as described under Materials and Methods. (C) The mRNA expression levels of RUNX2, OCN, OSX, and CEMP-1 were assessed by real-time reverse transcription (qRT)-PCR after 7 days of osteogenic induction. The values are shown as the mean ± standard deviation (n = 5). * p < 0.05 and ** p < 0.001 vs. the control value. # p < 0.05 and ## p < 0.001 vs. the Met-CM value at each time point.
Jfb 16 00177 g003
Figure 4. RNA sequencing of periodontal ligament stem cells (PDLSCs) cultured with osteogenic medium, osteogenic medium + lipopolysaccharide (LPS), metformin-preconditioned PDLSC medium (Met-CM), or LPS + Met-CM. (A) Heat map of significantly differentially expressed genes (DEGs) (fold-change ≥2 and p < 0.05). (B) Numbers of up- and downregulated DEGs in PDLSCs. Red indicates upregulation, and blue indicates downregulation. (C) Functional gene categories in each comparison.
Figure 4. RNA sequencing of periodontal ligament stem cells (PDLSCs) cultured with osteogenic medium, osteogenic medium + lipopolysaccharide (LPS), metformin-preconditioned PDLSC medium (Met-CM), or LPS + Met-CM. (A) Heat map of significantly differentially expressed genes (DEGs) (fold-change ≥2 and p < 0.05). (B) Numbers of up- and downregulated DEGs in PDLSCs. Red indicates upregulation, and blue indicates downregulation. (C) Functional gene categories in each comparison.
Jfb 16 00177 g004
Figure 5. Top 10 Gene Ontology (GO) terms from the biological processes and molecular functions enriched among differentially expressed genes (DEGs) identified from the (A) control vs. metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) and (B) lipopolysaccharide (LPS) vs. LPS + Met-CM comparisons using GO analysis in DAVID (p < 0.05).
Figure 5. Top 10 Gene Ontology (GO) terms from the biological processes and molecular functions enriched among differentially expressed genes (DEGs) identified from the (A) control vs. metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) and (B) lipopolysaccharide (LPS) vs. LPS + Met-CM comparisons using GO analysis in DAVID (p < 0.05).
Jfb 16 00177 g005
Figure 6. Image of the inositol phosphate metabolism pathway enriched among the imported DEGs identified from the control vs. metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) comparison. The red stars indicate key genes.
Figure 6. Image of the inositol phosphate metabolism pathway enriched among the imported DEGs identified from the control vs. metformin-preconditioned periodontal ligament stem cell (PDLSC) medium (Met-CM) comparison. The red stars indicate key genes.
Jfb 16 00177 g006
Figure 7. Images of the protein processing in the endoplasmic reticulum pathways enriched among the imported differentially expressed genes (DEGs) identified from the lipopolysaccharide (LPS) vs. LPS + Met-CM comparison. The red stars indicate key genes.
Figure 7. Images of the protein processing in the endoplasmic reticulum pathways enriched among the imported differentially expressed genes (DEGs) identified from the lipopolysaccharide (LPS) vs. LPS + Met-CM comparison. The red stars indicate key genes.
Jfb 16 00177 g007
Figure 8. Protein–protein interaction (PPI) network analysis using the STRING database. (A) The MAPK3 and CCNA2 hub networks for differentially expressed genes (DEGs) identified from the control vs. metformin-preconditioned medium (Met-CM) comparison. (B) The AKT1 and CTNNB1 hub networks for DEGs identified from the lipopolysaccharide (LPS) vs. LPS + Met-CM comparison. Each node marks a protein, and each edge designates an interaction. A confidence (interaction) score of at least 0.4 was set for significance. The PPI network was organized and visualized using Cytoscape v3.10.0.
Figure 8. Protein–protein interaction (PPI) network analysis using the STRING database. (A) The MAPK3 and CCNA2 hub networks for differentially expressed genes (DEGs) identified from the control vs. metformin-preconditioned medium (Met-CM) comparison. (B) The AKT1 and CTNNB1 hub networks for DEGs identified from the lipopolysaccharide (LPS) vs. LPS + Met-CM comparison. Each node marks a protein, and each edge designates an interaction. A confidence (interaction) score of at least 0.4 was set for significance. The PPI network was organized and visualized using Cytoscape v3.10.0.
Jfb 16 00177 g008
Table 1. KEGG pathway enrichment analysis.
Table 1. KEGG pathway enrichment analysis.
IDTermCount Genep Value
Control vs.
Met-CM
hsa05225Hepatocellular carcinoma127.82 × 10−4
hsa05142Chagas disease90.001
hsa05200Pathways in cancer220.004
hsa05226Gastric cancer100.004
hsa00562Inositol phosphate metabolism70.004
hsa00600Sphingolipid metabolism60.005
hsa05216Thyroid cancer50.007
hsa05217Basal cell carcinoma60.010
hsa05221Acute myeloid leukemia60.013
hsa05417Lipid and atherosclerosis110.015
LPS vs.
LPS + Met-CM
hsa04932Non-alcoholic fatty liver disease612.45 × 10−10
hsa04141Protein processing in endoplasmic reticulum653.25 × 10−10
hsa05012Parkinson disease887.75 × 10−10
hsa05014Amyotrophic lateral sclerosis1118.69 × 10−10
hsa04144Endocytosis832.57 × 10−9
hsa05208Chemical carcinogenesis—reactive oxygen species762.96 × 10−9
hsa05020Prion disease883.29 × 10−9
hsa05016Huntington disease956.23 × 10−9
hsa05022Pathways of neurodegeneration—multiple diseases1321.58 × 10−8
hsa05210Colorectal cancer382.67 × 10−8
Table 2. Top hub proteins with the highest number of PPIs.
Table 2. Top hub proteins with the highest number of PPIs.
Gene NameProtein DescriptionNo. of Interacting Proteins
Control vs. Met-CM
MAPK3mitogen-activated protein kinase 323
CCNA2cyclin A220
MRPL4mitochondrial ribosomal protein L420
UTP18UTP18, small subunit processome component16
NIP7NIP7, nucleolar pre-rRNA processing protein16
LPS vs. LPS + Met-CM
ACTBactin, beta464
GAPDH glyceraldehyde-3-phosphate dehydrogenase413
AKT1 AKT1 substrate 1409
CTNNB1catenin beta 1352
UBA52ubiquitin A-52 residue ribosomal protein fusion product 1345
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Suh, H.N.; Ji, J.Y.; Heo, J.S. Metformin-Enhanced Secretome from Periodontal Ligament Stem Cells Promotes Functional Recovery in an Inflamed Periodontal Model: In Vitro Study. J. Funct. Biomater. 2025, 16, 177. https://doi.org/10.3390/jfb16050177

AMA Style

Suh HN, Ji JY, Heo JS. Metformin-Enhanced Secretome from Periodontal Ligament Stem Cells Promotes Functional Recovery in an Inflamed Periodontal Model: In Vitro Study. Journal of Functional Biomaterials. 2025; 16(5):177. https://doi.org/10.3390/jfb16050177

Chicago/Turabian Style

Suh, Han Na, Ju Young Ji, and Jung Sun Heo. 2025. "Metformin-Enhanced Secretome from Periodontal Ligament Stem Cells Promotes Functional Recovery in an Inflamed Periodontal Model: In Vitro Study" Journal of Functional Biomaterials 16, no. 5: 177. https://doi.org/10.3390/jfb16050177

APA Style

Suh, H. N., Ji, J. Y., & Heo, J. S. (2025). Metformin-Enhanced Secretome from Periodontal Ligament Stem Cells Promotes Functional Recovery in an Inflamed Periodontal Model: In Vitro Study. Journal of Functional Biomaterials, 16(5), 177. https://doi.org/10.3390/jfb16050177

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop