Next Article in Journal
Effect of Cylinder-by-Cylinder Variation on Performance and Gaseous Emissions of a PFI Spark Ignition Engine: Experimental and 1D Numerical Study
Next Article in Special Issue
Doxycycline-Loaded Nitric Oxide-Releasing Nanomatrix Gel in Replanted Rat Molar on Pulp Regeneration
Previous Article in Journal
Highland Barley Replaces Sorghum as Raw Material to Make Shanxi Aged Vinegar
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Characterization of Periodontal Ligament Stem Cells Derived from Supernumerary Teeth in Three-Dimensional Culture Method

1
Department of Pediatric Dentistry, Graduate School, Kyung Hee University, Seoul 02447, Korea
2
Department of Pediatric Dentistry, School of Dentistry, Kyung Hee University, Seoul 02447, Korea
3
Department of Pediatric Dentistry, Kyung Hee University Dental Hospital, Seoul 02447, Korea
4
Department of Conservative Dentistry, Kyung Hee University School of Dentistry, Seoul 02447, Korea
*
Author to whom correspondence should be addressed.
Submission received: 11 May 2021 / Revised: 15 June 2021 / Accepted: 23 June 2021 / Published: 29 June 2021
(This article belongs to the Special Issue Applied Sciences in Oral Health and Clinical Dentistry)

Abstract

:
Objective: The aim of this study was to compare the characteristics of periodontal ligament stem cells derived from supernumerary teeth (sPDLSCs), cultured using a three-dimensional (3D) method and a conventional two-dimensional (2D) method. Methods: The morphology, viability, and osteogenic differentiation of the cells were analyzed. In addition, gene expression was analyzed by RNA sequencing, to characterize the functional differences. Results: The diameter of the 3D-cultured sPDLSCs decreased over time, but the spheroid shape was maintained for 7 days. The osteogenic differentiation was similar in the 2D and 3D. The gene expression related to the extracellular matrix (7.3%), angiogenesis (5.6%), cell proliferation (4.6%), inflammatory response (3.7%), and cell migration (3.5%) differed (p < 0.05). Conclusions: Within the limitations of this study, sPDLSCs varied in formation and function, depending on the culture method. In future, it is necessary to study tissue engineering using the advantages of 3D culture and the fewer ethical problems of supernumerary teeth.

1. Introduction

Mesenchymal stem cells (MSCs) can be harvested from various human tissues, such as bone marrow, adipose tissue, and umbilical cord blood [1]. In addition, MSCs have been extracted from the periodontal ligaments of permanent teeth, primary teeth, and supernumerary teeth [2,3,4]. Periodontal ligament (PDL) represents a fibrous network, connecting the cementum of the tooth root and the alveolar bone [5]. It serves many functions, such as tooth support, nutrition, and protection. Periodontal ligament stem cells (PDLSCs) are easy to obtain and have a greater proliferation rate than bone marrow stem cells. PDLSCs are capable of differentiating into osteoblasts, cementoblasts, adipocytes, and chondrocytes, and can form a structure similar to periodontal ligaments in vitro [6,7,8,9].
In 2011, PDLSCs were obtained from a supernumerary tooth (sPDLSCs) [4]. The sPDLSCs show a greater colony-forming ability than bone marrow stem cells, and can differentiate into adipocytes and osteoblasts [10]. Expendable dental tissue, such as supernumerary teeth, may be a source of stem cells. Therefore, studies of the biological properties of PDLSCs obtained from various dental tissue are needed to optimize their regenerative effects.
Cell culture is essential for the study of MSCs. In conventional two-dimensional (2D) cell culture methods, cells are cultured on the bottom of a culture-compatible polystyrene plate. Such cultures create an environment for studying biological processes or cell mechanisms under specific experimental conditions, and maintain various types of proliferation [11]. However, in nature, cells grow three-dimensionally (3D) rather than in a 2D plane. Therefore, it is difficult to study cell-to-cell signal transduction and the functions of cells in a conventional 2D culture environment [12]. To overcome these limitations, cell research using 3D culture is under investigation. Three-dimensional cultures have the advantage of emulating the original characteristics of the cell’s environment, so that the cell can display form, function, and activity as close to naturally as possible [13,14]. In addition, 3D culture is an alternative to animal experimentation [15].
There are the following two general methods of 3D culture: scaffold and scaffold-free. The scaffold method involves creating a scaffold of microstructures that the cells can use to move, proliferate, differentiate, and remain in contact [15]. However, scaffolds have limitations that can negatively affect cell stability and behavior during incubation [16]. In the scaffold-free method, a pallet or centrifugation tube culture, low-adhesion plate culture, bioreactor, hanging drop culture, or pallet culture and liquid overlay are used to create 3D microstructures without scaffolding [11,15,16,17]. In these methods, the cells assemble an endogenous extracellular matrix to form their own preferred microenvironment [18,19].
Cells cultured in a 3D environment behave differently than those in a 2D environment [17]. For example, primary articular chondrocytes and hepatocytes cultured in a 2D culture rapidly lose their normal phenotypes, but in 3D cultures the normal phenotype is maintained [20]. In addition, 3D-cultured stem cells exhibit better osteoblastic, adipogenic, and neuronal differentiation [21]. Similarly, 3D-cultured human dental pulp cells show higher expression of osteocalcin, dentin sialophosphoprotein, and alkaline phosphatase [22]. The expression of these proteins is influenced by cell–cell and cell–matrix interactions, which are difficult to imitate in 2D culture. In addition, the expression of genes related to multi-lineage differentiation, such as Nanog and Oct4, as well as those related to pluripotency transcription factors, is higher in 3D-cultured cells. These studies demonstrate that cell culture conditions influence gene expression [17,23]. However, previous studies have usually used microarrays and polymerase chain reaction, and few studies have analyzed genome-wide expression.
The aim of this study was to compare the characteristics of sPDLSCs cultured using a 3D and 2D method. Morphology, viability, osteogenic differentiation, and gene expression were evaluated. These experiments would reveal what physical and functional properties the same cells would have in different culture systems.

2. Materials and Methods

2.1. 2D and 3D Culture of Human sPDLSCs

sPDLSCs were purchased from Cell Engineering for Origin (Seoul, Korea). They were cultured in a 37 °C incubator with an atmosphere containing 5% CO2 in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Waltham, MA, USA). After three to four passages, the cells were split and thereafter grown in 2D or 3D cultures.
The formation of stem cell spheroids in 3D culture was performed with Stemfit 3D (Prosys Stemfit 3D®, Prodizen, Seoul, Korea; Figure 1). Cells (1.2 × 106) were seeded in a concave micromold and cultured in DMEM in a 37 °C incubator with an atmosphere containing 5% CO2.

2.2. Morphology

The 2D-cultured cells and the spheroids formed in 3D culture were observed at 1, 3, 5, and 7 days under a fluorescence microscope (JuLI; Nanoentek, Seoul, Korea).

2.3. Assays of Cell Viability

The cell counting kit-8 assay (CCK-8; Dojindo Molecular Technologies, Kumamoto, Japan) and live/dead viability/cytotoxicity kit for mammalian cells (Invitrogen, Carlsbad, CA, USA) were used to quantify cell proliferative ability. The CCK-8 assay is based on the activation of 1-methoxy phenazinium methylsulfate by nicotinamide adenine dinucleotide phosphate (NADP) and nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) produced by dehydrogenase in living cells. Water-soluble tetrazolium salt, 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H tetrazolium, and monosodium salt (WST-8) are reduced to orange formazan; therefore, the amount of WST-8 formazan produced by dehydrogenases is directly proportional to the number of viable cells.
The live/dead assay quantifies living and dead cells by measuring their esterase activity and plasma membrane integrity. The non-fluorescent dye calcein AM is converted into a fluorescent calcein that fluoresces in the presence of esterase in living cells, producing a uniform green fluorescence. Ethidium homodimer-1 enters dead cells through damaged membranes and binds with nucleic acids to produce red fluorescence.
The 2D- and 3D-cultured sPDLSCs were seeded at a density of 1.0 × 105 per well on a six-well plate and subjected to live/dead assay on days 3, 5, and 7. Images were obtained with a fluorescence microscope (IX71; Olympus, Tokyo, Japan). CCK-8 assay was performed on days 1, 3, 5, and 10, and the absorbance at 450 nm was measured with a Benchmark Plus multiplate spectrophotometer (Bio-Rad, Hercules, CA, USA).

2.4. Osteogenic Differentiation

After 2D or 3D culture, sPDLSCs were cultured in osteo-inductive medium (osteo group) or basal medium (control group) to confirm their ability to differentiate into bone cells. The osteo group was cultured for 2 weeks in osteo-inductive medium with 15% fetal bovine serum, 250 µL gentamycin reagent solution (final concentration 5 µg/mL), 5 mL filtered L-ascorbic acid (final concentration 10 mM), and 5 mL filtered dexamethasone. Alizarin red S (ARS) staining was performed to visualize calcium formation for evaluating differentiation. Cells were fixed in ice-cold 70% ethanol for 15 min, stained with ARS solution for 3 min, washed, and observed under an optical microscope. For quantitative evaluation of hard tissue formation, 10% cetylpyridinium chloride was added for 10 min, and the ARS stain was extracted and transferred to a 96-well plate. The absorbance was measured at a wavelength of 562 nm with a Benchmark Plus multiplate spectrophotometer (Bio-Rad).

2.5. Gene Expression

2.5.1. RNA Isolation

RNA was isolated from 2D- and 3D-cultured sPDLSCs on day 5 with RiboEx (GeneAll, Seoul, Korea), and RNA quality was assessed with an Agilent 2100 bioanalyzer with the RNA 6000 nano chip (Agilent Technologies, Amstelveen, The Netherlands). RNA quantification was performed with an ND-2000 spectrophotometer (Thermo, Waltham, DE, USA).

2.5.2. Library Preparation and Sequencing

Libraries of control and test RNA were constructed with the QuantSeq 3′ mRNA-Seq library prep kit (Lexogen, Vienna, Austria) according to the manufacturer’s instructions. In brief, 500 ng RNA was prepared, hybridized to an oligo-dT primer containing an Illumina-compatible sequence at its 5′ end, and subjected to reverse transcription. After degradation of the RNA template, second-strand synthesis was initiated with a random primer containing an Illumina-compatible linker sequence at its 5′ end. The double-stranded library was purified with magnetic beads to remove all reaction components. Next the library was amplified to add the complete adapter sequences required for cluster generation. The finished library was purified to remove PCR components. High-throughput sequencing was performed as single-end 75 bp sequencing using a NextSeq 500 sequencer (Illumina, San Diego, CA, USA).

2.5.3. Data Processing for the Identification of Differentially Expressed Genes

QuantSeq 3′ mRNA-Seq reads were aligned with Bowtie 2. Bowtie 2 indices were generated from a genome assembly sequence or the representative transcript sequences for aligning the genome and transcriptome. The alignment file was used to assemble transcripts, estimate their abundance, and detect the differential expression of genes. Differentially expressed genes were identified based on counts from unique and multiple alignments using coverage in BEDtools. The read count data were processed based on the quantile normalization method with EdgeR in R (R Development Core Team, Vienna, Austria) with Bioconductor.

2.6. Statistical Analyses

Statistical analyses were performed with SPSS version 25.0 (IBM, Armonk, NY, USA). One-way analysis of variance (ANOVA) and the Scheffé post hoc test were performed to compare absorbance values. p < 0.05 was considered indicative of statistical significance.

3. Results

3.1. Morphology

The sPDLSCs grown in the 2D culture exhibited a bipolar and stellate form, were attached to the plate, and increased in number over time (Figure 2A–D). The sPDLSCs grown in the 3D culture aggregated and became spheroid for about 24 h (Figure 2E). Although the diameters of the spheroids decreased significantly over the first 5 days (75.3%), they maintained their shape (Figure 2E–H).

3.2. Cell Viability

The absorbance of 2D-cultured sPDLSCs increased significantly over time, from 0.26 to 2.60 (Figure 3). In particular, it increased exponentially on day 10 (p < 0.05). By contrast, the absorbance of 3D-cultured sPDLSCs decreased over time, from 0.31 to 0.25. This decrease was significant from days 3 to 5 (p < 0.05), but not between days for the first 3 days (p > 0.05). At day 10, the absorbance increased slightly, but not significantly (p > 0.05).
The 2D-cultured sPDLSCs did not show a significant decrease in viability; however, the cell viability decreased significantly in the center of the spheroids in the 3D culture (Figure 4).

3.3. Stemness and Osteogenic Differentiation

ARS staining revealed significant extracellular calcium deposits in both the 2D and 3D cultures, which was indicative of osteogenic differentiation (Figure 5). Compared to the control group, the absorbance of the ARS stain extract was 2.3 in the 3D group, which was slightly higher than in the 2D group.

3.4. Identification of Differentially Expressed Genes and Gene Ontology Analyses

The expression of 25,737 differentially expressed genes in the 2D- and 3D-cultured sPDLSCs was examined. Of these, 5664 genes, related to cell function, immunity, inflammation, and the extracellular matrix, were selected for further analyses. The expression of 89 genes (58.9%) was upregulated, and that of 62 genes (41.1%) was downregulated in the 3D culture compared to in the 2D culture. The genes up- or downregulated were related to the extracellular matrix (ECM; 7.3%), angiogenesis (5.6%), cell proliferation (4.6%), inflammatory response (3.7%), and cell migration (3.5%; Figure 6). The genes in the 3D-cultured sPDLSCs that showed at least a 50-fold increase or decrease in expression compared to the 2D-cultured sPDLSCs are listed in Table 1 and Table 2.

4. Discussion

A scaffold-free 3D culture method, Stemfit 3D®, was used for the 3D culture of sPDLSCs, and was compared to a traditional 2D culture. The analyses of morphology, viability, osteogenic differentiation, and gene expression confirmed that the functions and characteristics of the sPDLSCs varied depending on the culture method.
Stemfit 3D® uses a non-adhesive plate, which is 600 µm in diameter, with a polydimethylsiloxane-based concave micromold. Concave micromolds were used because they accelerate cell aggregation compared to other methods (plane, cylindrical), and the cells form spheroids of uniform size that are easily harvested [2]. Moreover, 3D culture methods that use low-adhesive plates have lower long-term stability than other methods, because of the difficulty limiting the size of the spheroids [21]. However, because Stemfit 3D® can control the size of the spheroids, it shows improved stability over a longer period. Because of its economy, its ease of use, and the stability of the spheroids, Stemfit 3D® is commonly used in stem cell cytology and studies of other types of cells [3,4].
The diameter of the sPDLSC spheroids in the 3D culture decreased (75.3%) over the first 5 days, and decreased more slowly thereafter. The initial reduction in diameter was caused by cell aggregation and organization, but the subsequent reduction appears to have been a result of restricted nutrients and oxygen. Several models for the transport of nutrients, oxygen, and waste in spheroids have been verified [8]. When the diffusion of oxygen is limited, avascular tissue forms, and inefficient transport results in the accumulation of metabolic waste and the formation of a necrotic core at the center of the spheroid [10,12,14]. Several studies on MSC spheroids, including ones by Hildebrandt et al. [14], who investigated human MSCs, and by Yamaguchi et al. [18], who evaluated rat MSCs, have reported that spheroids form within 1 day and that their diameters decrease over time. However, Lee et al. [19] reported that the diameter of human dental pulp stem cell spheroids, cultured on a six-well non-adhesive plate, increased over time. This discrepancy is due to the different culture methods used. Hildebrandt et al. [14] and Yamaguchi et al. [18] independently cultured MSCs in a small volume of medium, or used the scaffold method, whereas Lee et al. [19] used a relatively large volume of medium. It is possible that spheroid–spheroid interactions may increase the diameter of the spheroids, but further study is needed.
CCK-8 and live/dead assays were used to confirm the proliferative ability and viability of 3D-cultured sPDLSCs; these are frequently used in 2D culture experiments [22,23]. The CCK-8 assay revealed that the absorbance of the 2D culture increased over time, and that of 3D culture tended to decrease over time. This was probably due to continued proliferation of cells in the 2D environment, whereas in the 3D environment the differentiation rate decreased as a result of restricted nutrients and oxygen in the center of the spheroids [20]. In addition, in the 3D culture, the number of dead cells in the center of the spheroids increased slightly over time. Similarly, in one study, spheroids of human MSCs in pallet culture maintained a stable internal structure for one month. However, a necrotic area developed in the center of the spheroids, with a loss of proliferation, impaired structural stability, and decreased cell-to-cell contact at two months [20]. Although quantitatively evaluating 2D and 3D culture methods is difficult [24], these results support the notion that the characteristics of 3D-cultured cells are fundamentally different from those of 2D-cultured cells.
Three-dimensional culture improves osteogenic differentiation compared to 2D culture. In addition, the intercellular interactions of embryonic stem cells are enhanced in a 3D environment [25]. For example, progenitor cells derived from the salivary gland can differentiate into hepatocytic and pancreatic islet cell lineages only when cultured in a 3D environment [26]. For this reason, 3D culture has attracted attention in stem cell biology and oncology, as well as in dentistry [12]. For example, 3D-cultured dental pulp stem cells or gingiva/papilla-derived stem cells show higher ALP activity than those grown in 2D culture [3,6,7]. In addition, the expression of genes associated with bone formation, such as BMP2, RUNX2, and dentine sialophosphoprotein, is increased in 3D-cultured cells [19].
The expression of BMP2 was upregulated 112.8-fold in 3D culture compared to in 2D culture. However, compared to our previous studies, the expression of osteogenic differentiation factors, such as RUNX2 and BMP2, tended to be lower in sPDLSCs than in pPDLSCs. Therefore, sPDLSCs can differentiate in 2D and 3D cultures, but their osteogenic differentiation is lower than that of cells extracted from permanent teeth.
RNA sequencing was conducted to analyze the gene expression profiles of sPDLSCs grown in 2D and 3D cultures. Analyses of gene expression profiles by microarray or PCR are limited by the fact that only selected genes can be identified. Because RNA sequencing captures a wider range of active genes, it is possible to detect small changes in expression with a limited amount of transcript [27].
The RNA sequencing results showed that the expression of genes related to the ECM, angiogenesis, cell proliferation, and the inflammatory response (such as AREG, SMOC1, MMP1, STC1, MMP10, PGF, and PTGS2) was very high. Indeed, 3D-cultured MSCs reportedly exhibit increased expression of genes related to angiogenesis, inflammation, and proliferation, such as TGS-6, IL24, VEGF, FGF-2, CXCR4, MCP3, RANTES, EGF, and SDF [21]. The increased expression of angiogenic genes is confirmed by the observation that 3D-cultured MSCs had greater angiogenic effects than 2D-cultured MSCs, enhancing tissue regeneration [28]. In addition, 3D-cultured MSCs show increased expression of genes related to osteogenesis and adipogenesis, such as RUNX-2, BMP2, OXC, OC, OPN, COLI, OCN, DDPP, DMP1, LPL, FABP4, PLN4, and PPARr [15,18,19]. These genes promote adipogenic and osteogenic differentiation by increasing the production of ECM proteins and endogenous growth factors.
The initial coalescence of spheroids is caused by caspase-dependent IL-1 autocrine signaling, which upregulates EGR2 [16]. In this study, the 3D-cultured sPDLSCs showed upregulated expression of EGR2 (2.3-fold). In addition, expression of EGR1 and EGR3 increased 5.4- and 9.7-fold, respectively. Similarly, immunomodulators, such as TNFAIP6 (24.5-fold), are reportedly upregulated in response to IL1 autocrine signaling in 3D spheroids [29,30]. In the center of spheroids, where the apoptosis zone is formed as a result of nutrient and oxygen limitation, hypoxia- and apoptosis-related genes, such as VEGFA and hypoxia-inducible factor (HIF), are upregulated. In this study, hypoxia-associated genes, such as VEGFA (6.1-fold) and HIF3A (2.0-fold), and apoptosis-related genes, such as IL24 (156-fold) and COMP (SFRP2), were highly upregulated in the spheroids. In addition, the expression of genes related to angiogenesis, such as PGF (341-fold) and PTGS2 (195.6-fold), was highly upregulated. These results are consistent with the observation that over time, the apoptotic zone of the spheroid increased and the surface proliferation zone decreased.
This study has several limitations. First, the decreased diameter and reduced proliferative ability of the 3D-cultured sPDLSCs may have been a result of their inherent characteristics or the morphologic characteristics of the spheroid, which hamper the supply of nutrients and oxygen, and the discharge of waste products. Our results are compatible with those of other studies that used 3D pallet cultures, low-adhesive plates, or scaffolds. In this regard, it may be necessary to compare other 3D methods, such as a bioreactor. Second, it may not have been optimal to use assays optimized for a 2D environment to evaluate 3D-cultured cells. For example, the CCK-8 assay involves measuring the color change caused by the dehydrogenation of water-soluble tetrazolium salts by living cells. Therefore, it is not ideal for comparing the absorbance of 2D and 3D cultures quantitatively, because the number of cells contacting the solution differs. It is difficult, or impossible, to find protocols and assays optimized for 3D-cultured sPDLSCs, so further research is needed to establish optimized methods for analyzing 3D-cultured cells. Third, this was a pilot study of 2D- and 3D-cultured sPDLSCs, and so the sample was small. Fourth, several prior studies on spheroid morphology involved observations over only 5–7 days. In the previous work of this research team, pPDLSCs were observed for 20 days to confirm their long-term stability [9]. However, the long-term stability of sPDLSC spheroids was not evaluated in this study. Therefore, further investigation of the long-term stability of sPDLSC spheroids is needed.
We found that 3D-cultured sPDLSCs exhibit a different gene expression profile, morphology, and physiology compared to identical 2D-cultured cells. Our results contribute to the development of optimized methods for the 3D culture of dental stem cells.

5. Conclusions

Supernumerary teeth, as expendable dental tissue, may be ethically less restrictive for clinical use than permanent teeth. Thus, they could be a good source of stem cells. Within the limitations of this study, we confirmed that the function of sPDLSCs varies depending on the culture method used. In addition, 3D-cultured sPDLSCs have greater stemness than 2D-cultured sPDLSCs. Our findings accelerate the development of regenerative medicine using MSCs derived from expendable tissue, including supernumerary teeth. Further studies that aim to enable the clinical application of 3D-cultured sPDLSCs are warranted.

Author Contributions

Conceptualization, Y.Y.J. and H.-S.L.; methodology, J.-H.J.; software, M.S.K.; validation, Y.Y.J., H.-S.L. and J.-H.J.; formal analysis, K.E.L.; investigation, O.H.N.; resources, S.-C.C.; data curation, Y.Y.J.; writing—original draft preparation, Y.Y.J. and H.-S.L.; writing—review and editing, M.S.K., K.E.L., O.H.N., J.-H.J. and S.-C.C.; visualization, Y.Y.J.; supervision, H.-S.L.; project administration, H.-S.L.; funding acquisition, H.-S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation of Korea (NRF-No.2019R1G1A1100082) and National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No.2021R1G1A1013927).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are openly available.

Acknowledgments

We would like to thank Kwon Il-Geun who greatly supported this research.

Conflicts of Interest

The authors declare no conflict of interest.

Summary of Abbreviations

AbbreviationsDefinition
2DTwo-dimensional
3DThree-dimensional
ARSAlizarin red S
ECMExtracellular matrix
HIFHypoxia inducible factor
MSCsMesenchymal stem cells
NADPNicotinamide adenine dinucleotide phosphate
NADPHNicotinamide adenine dinucleotide phosphate hydrogen
PDLPeriodontal ligament
PDLSCsPeriodontal ligament stem cells
sPDLSCsperiodontal ligament stem cells derived from supernumerary teeth

References

  1. Jin, H.J.; Bae, Y.K.; Kim, M.; Kwon, S.J.; Jeon, H.B.; Choi, S.J.; Kim, S.W.; Yang, Y.S.; Oh, W.; Chang, J.W. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int. J. Mol. Sci. 2013, 14, 17986–18001. [Google Scholar] [CrossRef]
  2. Wong, S.F.; da No, Y.; Choi, Y.Y.; Kim, D.S.; Chung, B.G.; Lee, S.H. Concave microwell based size-controllable hepatosphere as a three-dimensional liver tissue model. Biomaterials 2011, 32, 8087–8096. [Google Scholar] [CrossRef]
  3. Lee, S.I.; Ko, Y.; Park, J.B. Evaluation of the maintenance of stemness, viability, and differentiation potential of gingiva-derived stem-cell spheroids. Exp. Ther. Med. 2017, 13, 1757–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kim, Y.; Baipaywad, P.; Jeong, Y.; Park, H. Incorporation of gelatin microparticles on the formation of adipose-derived stem cell spheroids. Int. J. Biol. Macromol. 2018, 110, 472–478. [Google Scholar] [CrossRef] [PubMed]
  5. Di Vito, A.; Giudice, A.; Chiarella, E.; Malara, N.; Bennardo, F.; Fortunato, L. In Vitro Long-Term Expansion and High Osteogenic Potential of Periodontal Ligament Stem Cells: More Than a Mirage. Cell Transpl. 2019, 28, 129–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lee, S.I.; Ko, Y.; Park, J.B. Evaluation of the shape, viability, stemness and osteogenic differentiation of cell spheroids formed from human gingiva-derived stem cells and osteoprecursor cells. Exp. Ther. Med. 2017, 13, 3467–3473. [Google Scholar] [CrossRef]
  7. Lee, S.I.; Yeo, S.I.; Kim, B.B.; Ko, Y.; Park, J.B. Formation of size-controllable spheroids using gingiva-derived stem cells and concave microwells: Morphology and viability tests. Biomed. Rep. 2016, 4, 97–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Jiang, Y.; Pjesivac-Grbovic, J.; Cantrell, C.; Freyer, J.P. A multiscale model for avascular tumor growth. Biophys. J. 2005, 89, 3884–3894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Jeong, Y.Y.; Kim, M.S.; Lee, K.E.; Nam, O.H.; Jang, J.-H.; Choi, S.-C.; Lee, H.-S. Comparison of 2- and 3-Dimensional Cultured Periodontal Ligament Stem Cells. Pilot Study 2021, 11, 1083. [Google Scholar]
  10. Curcio, E.; Salerno, S.; Barbieri, G.; De Bartolo, L.; Drioli, E.; Bader, A. Mass transfer and metabolic reactions in hepatocyte spheroids cultured in rotating wall gas-permeable membrane system. Biomaterials 2007, 28, 5487–5497. [Google Scholar] [CrossRef]
  11. Mueller-Klieser, W. Method for the determination of oxygen consumption rates and diffusion coefficients in multicellular spheroids. Biophys. J. 1984, 46, 343–348. [Google Scholar] [CrossRef]
  12. Cesarz, Z.; Tamama, K. Spheroid Culture of Mesenchymal Stem Cells. Stem Cells Int. 2016, 2016, 9176357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lin, R.Z.; Chang, H.Y. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol. J. 2008, 3, 1172–1184. [Google Scholar] [CrossRef]
  14. Hildebrandt, C.; Buth, H.; Thielecke, H. A scaffold-free in vitro model for osteogenesis of human mesenchymal stem cells. Tissue Cell 2011, 43, 91–100. [Google Scholar] [CrossRef] [PubMed]
  15. Yamamoto, M.; Kawashima, N.; Takashino, N.; Koizumi, Y.; Takimoto, K.; Suzuki, N.; Saito, M.; Suda, H. Three-dimensional spheroid culture promotes odonto/osteoblastic differentiation of dental pulp cells. Arch. Oral Biol. 2014, 59, 310–317. [Google Scholar] [CrossRef]
  16. Yeh, H.Y.; Liu, B.H.; Sieber, M.; Hsu, S.H. Substrate-dependent gene regulation of self-assembled human MSC spheroids on chitosan membranes. BMC Genom. 2014, 15, 10. [Google Scholar] [CrossRef] [Green Version]
  17. Kikuchi, H.; Suzuki, K.; Sakai, N.; Yamada, S. Odontoblasts induced from mesenchymal cells of murine dental papillae in three-dimensional cell culture. Cell Tissue Res. 2004, 317, 173–185. [Google Scholar] [CrossRef]
  18. Yamaguchi, Y.; Ohno, J.; Sato, A.; Kido, H.; Fukushima, T. Mesenchymal stem cell spheroids exhibit enhanced in-vitro and in-vivo osteoregenerative potential. BMC Biotechnol. 2014, 14, 105. [Google Scholar] [CrossRef] [Green Version]
  19. Lee, S.H.; Inaba, A.; Mohindroo, N.; Ganesh, D.; Martin, C.E.; Chugal, N.; Kim, R.H.; Kang, M.K.; Park, N.H.; Shin, K.H. Three-dimensional Sphere-forming Cells Are Unique Multipotent Cell Population in Dental Pulp Cells. J. Endod. 2017, 43, 1302–1308. [Google Scholar] [CrossRef]
  20. Bellotti, C.; Duchi, S.; Bevilacqua, A.; Lucarelli, E.; Piccinini, F. Long term morphological characterization of mesenchymal stromal cells 3D spheroids built with a rapid method based on entry-level equipment. Cytotechnology 2016, 68, 2479–2490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Sart, S.; Tsai, A.C.; Li, Y.; Ma, T. Three-dimensional aggregates of mesenchymal stem cells: Cellular mechanisms, biological properties, and applications. Tissue Eng. Part B Rev. 2014, 20, 365–380. [Google Scholar] [CrossRef] [Green Version]
  22. Ishiyama, M.; Miyazono, Y.; Sasamoto, K.; Ohkura, Y.; Ueno, K. A highly water-soluble disulfonated tetrazolium salt as a chromogenic indicator for NADH as well as cell viability. Talanta 1997, 44, 1299–1305. [Google Scholar] [CrossRef]
  23. Amirikia, M.; Ali Jorsaraei, S.G.; Ali Shariatzadeh, S.M.; Mehranjani, M.S. Differentiation of stem cells from the apical papilla into osteoblasts by the elastic modulus of porous silk fibroin scaffolds. Biologicals 2019, 57, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Fennema, E.; Rivron, N.; Rouwkema, J.; van Blitterswijk, C.; de Boer, J. Spheroid culture as a tool for creating 3D complex tissues. Trends Biotechnol. 2013, 31, 108–115. [Google Scholar] [CrossRef] [PubMed]
  25. Yan, X.Z.; van den Beucken, J.; Yuan, C.; Jansen, J.A.; Yang, F. Spheroid formation and stemness preservation of human periodontal ligament cells on chitosan films. Oral. Dis. 2018, 24, 1083–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Okumura, K.; Nakamura, K.; Hisatomi, Y.; Nagano, K.; Tanaka, Y.; Terada, K.; Sugiyama, T.; Umeyama, K.; Matsumoto, K.; Yamamoto, T.; et al. Salivary gland progenitor cells induced by duct ligation differentiate into hepatic and pancreatic lineages. Hepatology 2003, 38, 104–113. [Google Scholar] [CrossRef]
  27. Zhao, S.; Fung-Leung, W.P.; Bittner, A.; Ngo, K.; Liu, X. Comparison of RNA-Seq and microarray in transcriptome profiling of activated T cells. PLoS ONE 2014, 9, e78644. [Google Scholar] [CrossRef]
  28. Laschke, M.W.; Schank, T.E.; Scheuer, C.; Kleer, S.; Schuler, S.; Metzger, W.; Eglin, D.; Alini, M.; Menger, M.D. Three-dimensional spheroids of adipose-derived mesenchymal stem cells are potent initiators of blood vessel formation in porous polyurethane scaffolds. Acta Biomater. 2013, 9, 6876–6884. [Google Scholar] [CrossRef] [PubMed]
  29. Tsai, A.C.; Liu, Y.; Yuan, X.; Ma, T. Compaction, fusion, and functional activation of three-dimensional human mesenchymal stem cell aggregate. Tissue Eng. Part A 2015, 21, 1705–1719. [Google Scholar] [CrossRef] [Green Version]
  30. Bartosh, T.J.; Ylostalo, J.H.; Bazhanov, N.; Kuhlman, J.; Prockop, D.J. Dynamic compaction of human mesenchymal stem/precursor cells into spheres self-activates caspase-dependent IL1 signaling to enhance secretion of modulators of inflammation and immunity (PGE2, TSG6, and STC1). Stem Cells 2013, 31, 2443–2456. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Stemfit 3D® uses a non-adhesive plate with a polydimethylsiloxane-based concave micromold 600 µm in diameter. It can control the size of spheroids and enhance their long-term stability.
Figure 1. Stemfit 3D® uses a non-adhesive plate with a polydimethylsiloxane-based concave micromold 600 µm in diameter. It can control the size of spheroids and enhance their long-term stability.
Applsci 11 06040 g001
Figure 2. Morphology, days 1–7. (AD) Morphology of sPDLSCs at days 1, 3, 5, and 7 in 2D cultures (magnification, ×200). The sPDLSCs increased in number over time. (EH) Spheroid of sPDLSCs at days 1,3,5, and 7 in 3D cultures (magnification, ×200). The sPDLSCs aggregated and became spheroid for about 24h (E). Diameters of the spheroids decreased significantly over the time.
Figure 2. Morphology, days 1–7. (AD) Morphology of sPDLSCs at days 1, 3, 5, and 7 in 2D cultures (magnification, ×200). The sPDLSCs increased in number over time. (EH) Spheroid of sPDLSCs at days 1,3,5, and 7 in 3D cultures (magnification, ×200). The sPDLSCs aggregated and became spheroid for about 24h (E). Diameters of the spheroids decreased significantly over the time.
Applsci 11 06040 g002
Figure 3. Results of CCK-8 assays. The 2D-cultured sPDLSCs showed an increase in absorbance and 3D-cultured sPDLSCs showed a significant decrease in absorbance from day 1 to day 5. The absorbance of 3D-cultured sPDLSCs increased non-significantly on day 10. * p < 0.05; Scheffé post hoc test following one-way ANOVA.
Figure 3. Results of CCK-8 assays. The 2D-cultured sPDLSCs showed an increase in absorbance and 3D-cultured sPDLSCs showed a significant decrease in absorbance from day 1 to day 5. The absorbance of 3D-cultured sPDLSCs increased non-significantly on day 10. * p < 0.05; Scheffé post hoc test following one-way ANOVA.
Applsci 11 06040 g003
Figure 4. Live/dead assays. The 2D-cultured sPDLSCs did not show a significant decrease in viability; however, the cell viability decreased significantly in the center of the spheroids in the 3D culture. (AF) Live/dead assay results for 2D-cultured sPDLSCs at days 3, 5, and 7 (magnification, ×100). The number of cells increased, and no dead cells were observed. (GL) Live/dead assay results for 3D-cultured sPDLSCs at days 3, 5, and 7 (magnification, ×100). The number of dead cells in the center of spheroids increased with decreasing spheroid diameter.
Figure 4. Live/dead assays. The 2D-cultured sPDLSCs did not show a significant decrease in viability; however, the cell viability decreased significantly in the center of the spheroids in the 3D culture. (AF) Live/dead assay results for 2D-cultured sPDLSCs at days 3, 5, and 7 (magnification, ×100). The number of cells increased, and no dead cells were observed. (GL) Live/dead assay results for 3D-cultured sPDLSCs at days 3, 5, and 7 (magnification, ×100). The number of dead cells in the center of spheroids increased with decreasing spheroid diameter.
Applsci 11 06040 g004
Figure 5. Evaluation of osteogenesis in 2D- and 3D-cultured sPDLSCs (magnification, ×200). (A) Control group of 2D-cultured sPDLSCs in basal media. (B) Osteo group of 2D-cultured sPDLSCs in osteogenic media. ARS staining revealed significant extracellular calcium deposits (yellow circles). (C) Control group of 3D-cultured sPDLSCs in basal media. (D) Osteo group of 3D-cultured sPDLSCs in osteogenic media.
Figure 5. Evaluation of osteogenesis in 2D- and 3D-cultured sPDLSCs (magnification, ×200). (A) Control group of 2D-cultured sPDLSCs in basal media. (B) Osteo group of 2D-cultured sPDLSCs in osteogenic media. ARS staining revealed significant extracellular calcium deposits (yellow circles). (C) Control group of 3D-cultured sPDLSCs in basal media. (D) Osteo group of 3D-cultured sPDLSCs in osteogenic media.
Applsci 11 06040 g005
Figure 6. Expression of filtered gene categories. Distribution and ratio of genes in 10 selected categories with 10-fold increased expression (p < 0.05).
Figure 6. Expression of filtered gene categories. Distribution and ratio of genes in 10 selected categories with 10-fold increased expression (p < 0.05).
Applsci 11 06040 g006
Table 1. Genes upregulated more than 10-fold in 3D- compared to 2D-cultured sPDLSCs.
Table 1. Genes upregulated more than 10-fold in 3D- compared to 2D-cultured sPDLSCs.
Gene NameFold ChangeDescriptionRelated Function
AREG1172.7AmphiregulinCell differentiation, cell proliferation
SMOC11161.3SPARC-related modular calcium binding 1Extracellular matrix
MMP1829.2Matrix metallopeptidase 1Cell migration, extracellular matrix
STC1791.3Stanniocalcin 1Cell differentiation, cell proliferation
MMP10349.8Matrix metallopeptidase 10Extracellular matrix
PGF341.9Placental growth factorAngiogenesis
PTGS2195.6Prostaglandin-endoperoxide synthase 2Angiogenesis, cell differentiation, inflammatory response
COL5A3173.4Collagen type V alpha 3Extracellular matrix
IL24156.0Interleukin 24Apoptotic process, cell death
FRZB119.4Frizzled-related proteinCell differentiation
BMP2112.8Bone morphogenetic protein 2Cell differentiation, cell proliferation, inflammatory response
SFRP2104.5Secreted frizzled-related protein 2Angiogenesis, apoptotic process, cell death, cell differentiation, cell proliferation, extracellular matrix
FGL298.6Fibrinogen-like 2Immune response
IER397.3Immediate early response 3Apoptotic process, cell death
TFPI294.9Tissue factor pathway inhibitor 2Extracellular matrix
DPT83.6DermatopontinExtracellular matrix
NR4A282.5Nuclear receptor subfamily 4 group A member 2Cell differentiation, cell migration
TNC82.1Tenascin CCell differentiation, extracellular matrix
NPNT73.8NephronectinCell differentiation, extracellular matrix
NDP64.7Norrie disease (pseudoglioma)Cell proliferation, extracellular matrix
ACKR464.5Atypical chemokine receptor 4Immune response
IL18R163.5Interleukin 18 receptor 1Cell differentiation, immune response
MMP862.5Matrix metallopeptidase 8Extracellular matrix
SPRY161.6Sprouty RTK signaling antagonist 1Cell cycle, cell differentiation
RGS261.4Regulator of G-protein signaling 2Cell cycle, cell differentiation
IL1R160.3Interleukin 1 receptor type 1Immune response
PTGS160.3Prostaglandin-endoperoxide synthase 1Inflammatory response
SPON153.6Spondin 1Extracellular matrix
EDNRB52.4Endothelin receptor type BCell differentiation, cell migration
PECAM149.3Platelet and endothelial cell adhesion molecule 1Angiogenesis, cell differentiation, cell migration
IL1RN46.1Interleukin 1 receptor antagonistImmune response, inflammatory response
PRDM144.6PR domain 1Cell differentiation, cell proliferation, immune response
CHST242.1Carbohydrate sulfotransferase 2Inflammatory response
APOE40.0Apolipoprotein ECell differentiation, extracellular matrix
NOX438.8NADPH oxidase 4Cell differentiation, inflammatory response
SNAP2538.2Synaptosome-associated protein 25 kDaCell differentiation, immune response
TNN36.8Tenascin NCell differentiation, extracellular matrix
VAV334.8VAV guanine nucleotide exchange factor 3Angiogenesis, cell migration
BCL2A131.5BCL2-related protein A1Apoptotic process, cell death
JUP30.3Junction plakoglobinCell death, cell differentiation, cell migration, extracellular matrix, immune response
AKR1C130.3Aldo-keto reductase family 1, member C1Cell differentiation
NRN129.8Neuritin 1Cell differentiation
ITGA228.6Integrin subunit alpha 2Cell differentiation, cell migration, cell proliferation
SLC1A328.3Solute carrier family 1 member 3Cell differentiation
SECTM127.7Secreted and transmembrane 1Immune response
CD727.2CD7 moleculeImmune response
AKR1C227.1Aldo-keto reductase family 1, member C2Cell differentiation
AIM226.7Absent in melanoma 2Apoptotic process, cell death, immune response, inflammatory response
GPM6B26.5Glycoprotein M6BCell differentiation
ETV126.0ETS Variant 1Cell differentiation
CXCL824.7C-X-C motif chemokine ligand 8Angiogenesis, cell cycle, cell migration, immune response, inflammatory response
NKX2-524.5NK2 homeobox 5Apoptotic process, cell death, cell differentiation, cell proliferation
TNFAIP624.5TNF-alpha-induced protein 6Immune response, inflammatory response
NEFM23.6Neurofilament, medium polypeptideCell differentiation
CCL223.3C-C motif chemokine ligand 2Cell migration, immune response
KYNU22.7KynureninaseImmune response
RANBP3L22.5RAN-binding protein 3-likeCell cycle, cell differentiation
NTN121.6Netrin 1Apoptotic process, cell death, cell differentiation, cell migration, extracellular matrix
SEPT321.0Septin 3Cell cycle
GABRB220.4Gamma-aminobutyric acid type A receptor beta2 subunitCell differentiation
EDNRA20.3Endothelin receptor type AAngiogenesis, cell differentiation, cell proliferation
CHST119.5Carbohydrate sulfotransferase 1Inflammatory response
AFAP1L219.5Actin filament-associated protein 1-like 2Inflammatory response
CRYGD19.4Crystallin, gamma DCell differentiation
CMKLR118.6Chemerin chemokine-like receptor 1Immune response
WNT7B17.4Wnt family member 7BCell differentiation, cell proliferation, extracellular matrix
PTPRU16.5Protein tyrosine phosphatase, receptor type UCell differentiation
ITGB316.4Integrin subunit beta 3Angiogenesis, cell differentiation, cell migration
TNFRSF2115.5Tumor necrosis factor receptor superfamily member 21Apoptotic process, cell death, cell differentiation, immune response
NYAP114.4Neuronal tyrosine phosphorylated phosphoinositide-3-kinase adaptor 1Cell differentiation
GRIN2A13.3Glutamate ionotropic receptor NMDA type subunit 2ACell differentiation
PHOX2B12.8Paired-like homeobox 2bCell differentiation, cell migration
GPR6812.8G-protein-coupled receptor 68Inflammatory response
FPR112.7Formyl peptide receptor 1Cell migration, immune response, inflammatory response
EREG12.6EpiregulinAngiogenesis, cell cycle, cell differentiation, cell proliferation
FGF712.5Fibroblast growth factor 7Cell proliferation
TNFSF1012.1Tumor necrosis factor superfamily member 10Apoptotic process, cell death, immune response
LRP411.5LDL receptor-related protein 4Cell differentiation
PTPN2211.3Protein tyrosine phosphatase, non-receptor type 22Cell differentiation
RNF15211.3Ring finger protein 152Apoptotic process, cell death
SOCS211.3Suppressor of cytokine signaling 2Cell differentiation
CXCL311.2CXC motif chemokine ligand 3Cell migration, immune response, inflammatory response
GEM11.1GTP-binding protein overexpressed in skeletal muscleCell cycle, immune response
ZNF44310.8Zinc finger protein 443Apoptotic process, cell death
MGP10.7Matrix Gla proteinExtracellular matrix
TNIK10.5TRAF2 and NCK interacting kinaseCell differentiation
NR4A310.3Nuclear receptor subfamily 4 group A member 3Cell differentiation, cell proliferation, immune response
CSGALNACT110.3Chondroitin sulfate N-acetylgalactosaminyltransferase 1Cell proliferation
CTSS10.3Cathepsin SImmune response
Table 2. Genes downregulated >10-fold in 3D- compared to 2D-cultured sPDLSCs.
Table 2. Genes downregulated >10-fold in 3D- compared to 2D-cultured sPDLSCs.
Gene NameFold ChangeDescriptionRelated Function
ACAN177.4AggrecanCell differentiation, extracellular matrix
NGF136.7Nerve growth factorApoptotic process, cell death, cell differentiation
HAPLN1106.0Hyaluronan and proteoglycan link protein 1Extracellular matrix
NPR3102.8Natriuretic peptide receptor 3Cell proliferation
CTNND286.6Catenin delta 2Cell differentiation
ADM281.4Adrenomedullin 2Angiogenesis
TRIM6758.4Tripartite motif containing 67Cell differentiation
ADAMTS1455.6ADAM metallopeptidase with thrombospondin type 1 motif 14Extracellular matrix
DLGAP552.7Discs large homolog-associated protein 5Cell cycle, cell proliferation
ADRA1B45.1Adrenoceptor alpha 1BCell proliferation
LGR545.1Leucine-rich repeat-containing G-protein-coupled receptor 5Cell differentiation, cell proliferation
TENM241.3Teneurin transmembrane protein 2Cell differentiation
SLITRK141.2SLIT and NTRK like family member 1Cell differentiation
MAP238.3Microtubule associated protein 2Cell differentiation
IL7R35.5Interleukin 7 receptorCell differentiation, cell proliferation, immune response
GLI133.3GLI family zinc finger 1Cell differentiation
KRT33B32.6Keratin 33BCell death, cell differentiation
COL1A127.8Collagen type I alpha 1Cell differentiation, cell migration, extracellular matrix
HILS125.5Histone linker H1 domain, spermatid-specific 1 (pseudogene)Cell differentiation
ANK225.5Ankyrin 2, neuronalCell differentiation
PRELP25.0Proline/arginine-rich end leucine-rich repeat proteinCell differentiation, extracellular matrix
KRT3423.2Keratin 34Cell death, cell differentiation
TTK19.6TTK protein kinaseCell cycle
ASB218.7Ankyrin repeat and SOCS box-containing 2Cell differentiation
PSG117.8Pregnancy-specific beta-1-glycoprotein 1Cell migration
ASPN16.9AsporinExtracellular matrix
RSPO216.5R-Spondin 2Cell differentiation
TAGLN15.7TransgelinCell differentiation
FBN215.3Fibrillin 2Extracellular matrix
LOX15.1Lysyl oxidaseExtracellular matrix
SEMA3D14.4Semaphorin 3DCell differentiation
LEP14.0LeptinCell differentiation, cell migration
COL6A613.6Collagen type VI alpha 6Extracellular matrix
MMP1513.6Matrix metallopeptidase 15Extracellular matrix
CREB3L113.5cAMP-responsive element-binding protein 3-like 1Cell differentiation
EFHD113.5EF-hand domain family member D1Cell differentiation
KRTAP1-513.4Keratin-associated protein 1-5Cell differentiation
ANLN13.3Anillin actin-binding proteinCell cycle, cell differentiation, cell migration
HLA-DMA13.1Major histocompatibility complex, class II, DM alphaImmune response
MKI6713.0Marker of proliferation Ki-67Cell cycle, cell proliferation
GDF612.9Growth differentiation factor 6Cell death, cell differentiation
ANKRD112.7Ankyrin repeat domain 1Cell differentiation
BCL212.6B-cell CLL/lymphoma 2Cell death, cell differentiation, cell proliferation
RIMS112.5Regulating synaptic membrane exocytosis 1Cell differentiation
ECM212.4Extracellular matrix protein 2Cell differentiation, extracellular matrix
TUBB2B12.2Tubulin beta 2B class IIbCell differentiation, cell migration
CLDN112.0Claudin 1Cell differentiation, immune response
AURKB11.7Aurora kinase BCell cycle
RTN111.6Reticulon 1Cell differentiation
BCAT111.6Branched chain amino acid transaminase 1Cell cycle, cell proliferation
KIRREL311.5Kin of IRRE-like 3 (Drosophila)Cell differentiation, cell migration
SLC4A511.2Solute carrier family 4 member 5Cell differentiation
MYC11.1v-Myc avian myelocytomatosis viral oncogene homologCell cycle, cell differentiation, cell proliferation, immune response
CYR6111.0Cysteine-rich angiogenic inducer 61Angiogenesis, apoptotic process, cell death, cell differentiation, cell migration, cell proliferation, extracellular matrix
CCBE111.0Collagen and calcium-binding EGF domains 1Angiogenesis, extracellular matrix
TPM110.6Tropomyosin 1 (alpha)Cell differentiation
CHAC110.5ChaC glutathione-specific gamma-glutamylcyclotransferase 1Apoptotic process, cell death, cell differentiation
CTGF10.4Connective tissue growth factorAngiogenesis, cell differentiation, cell migration, cell proliferation, extracellular matrix
NAV210.3Neuron navigator 2Extracellular matrix
ETV710.2ETS Variant 7Cell differentiation
CLEC3B10.1C-type lectin domain family 3 member BExtracellular matrix
MATN210.1Matrilin 2Cell differentiation, cell migration, extracellular matrix
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jeong, Y.Y.; Kim, M.S.; Lee, K.E.; Nam, O.H.; Jang, J.-H.; Choi, S.-C.; Lee, H.-S. In Vitro Characterization of Periodontal Ligament Stem Cells Derived from Supernumerary Teeth in Three-Dimensional Culture Method. Appl. Sci. 2021, 11, 6040. https://0-doi-org.brum.beds.ac.uk/10.3390/app11136040

AMA Style

Jeong YY, Kim MS, Lee KE, Nam OH, Jang J-H, Choi S-C, Lee H-S. In Vitro Characterization of Periodontal Ligament Stem Cells Derived from Supernumerary Teeth in Three-Dimensional Culture Method. Applied Sciences. 2021; 11(13):6040. https://0-doi-org.brum.beds.ac.uk/10.3390/app11136040

Chicago/Turabian Style

Jeong, Yun Yeong, Mi Sun Kim, Ko Eun Lee, Ok Hyung Nam, Ji-Hyun Jang, Sung-Chul Choi, and Hyo-Seol Lee. 2021. "In Vitro Characterization of Periodontal Ligament Stem Cells Derived from Supernumerary Teeth in Three-Dimensional Culture Method" Applied Sciences 11, no. 13: 6040. https://0-doi-org.brum.beds.ac.uk/10.3390/app11136040

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