Abstract
Dental mesenchymal stem cells (DMSCs) are pivotal for tooth development and periodontal tissue health and play an important role in tissue engineering and regenerative medicine because of their multidirectional differentiation potential and self-renewal ability. The cellular microenvironment regulates the fate of stem cells and can be modified using various optimization techniques. These methods can influence the cellular microenvironment, activate disparate signaling pathways, and induce different biological effects. “Epigenetic regulation” refers to the process of influencing gene expression and regulating cell fate without altering DNA sequences, such as histone methylation. Histone methylation modifications regulate pivotal transcription factors governing DMSCs differentiation into osteo-/odontogenic lineages. The most important sites of histone methylation in tooth organization were found to be H3K4, H3K9, and H3K27. Histone methylation affects gene expression and regulates stem cell differentiation by maintaining a delicate balance between major trimethylation sites, generating distinct chromatin structures associated with specific downstream transcriptional states. Several crucial signaling pathways associated with osteogenic differentiation are susceptible to modulation via histone methylation modifications. A deeper understanding of the regulatory mechanisms governing histone methylation modifications in osteo-/odontogenic differentiation and immune-inflammatory responses of DMSCs will facilitate further investigation of the epigenetic regulation of histone methylation in DMSC-mediated tissue regeneration and inflammation. Here is a concise overview of the pivotal functions of epigenetic histone methylation at H3K4, H3K9, and H3K27 in the regulation of osteo-/odontogenic differentiation and renewal of DMSCs in both non-inflammatory and inflammatory microenvironments. This review summarizes the current research on these processes in the context of tissue regeneration and therapeutic interventions.
Bones and teeth are integral components of the craniofacial skeleton, providing indispensable support for the overlying soft tissues and allowing for complex and intricate movements.1 Missing teeth, due to congenital or acquired diseases, and trauma, is by far the most common oral health problem.2 Maxillofacial tumors can invade the normal tissue structures of the jaw, teeth, muscles, and skin, severely affecting maxillofacial form and function. The use of autologous or allogeneic bone grafts to repair maxillofacial bone defects caused by tumors or traumatic injuries is common, but problems such as poor osseointegration stability, immune rejection, and interference with normal physiological functions remain. In elderly individuals or those with severe periodontitis, severe damage to the dental tissue can result in malnutrition due to difficulty in eating and chewing, which may subsequently affect an individual’s self-esteem and lead to severe psychosocial and mental health issues.2 Prevailing techniques for the restoration of damaged and missing teeth rely on the use of synthetic materials to repair structural defects. However, these techniques do not facilitate the promotion of biological functions such as supplying blood and nerves.
Mesenchymal stem cells (MSCs) are a population of stem cells that function as self-renewing and differentiating progenitor cells. The International Society for Cell & Gene Therapy (ISCT) Mesenchymal Stromal Cell committee defines them as readily adherent, expressing CD105, CD73, and CD90 (but not CD11b, CD14, CD19, CD34, CD45, CD79a, HLA-DR). In addition, these cells are capable of differentiating into osteoblasts, chondroblasts, and adipocyte lineages in vitro.3,4 Bone marrow-derived mesenchymal stem cells (BM-MSCs) are a category of seed cells currently employed with considerable frequency in the field of organizational engineering.5,6 They are commonly used to repair bone defects owing to their ability to rapidly adapt to the body’s bone microenvironment and accelerate angiogenesis when implanted after mixing with scaffolding materials in vitro. DMSCs have been identified as an optimal cell source for regenerative therapies owing to their excellent proliferative capacity and multidirectional differentiation potential.7 Compared to BM-MSCs, DMSCs demonstrate augmented proliferative, neural differentiation, and odontogenic capabilities,8 which are highly promising for bone and neural tissue regeneration.9 The cellular microenvironment exerts a significant influence on the fate of stem cells and can be modified using various optimization techniques. These include hormone-stem cell combination therapy, gene modification to regulate stem cells, epigenetic regulation, cell membrane sheets, scaffolding materials, and hydrogels.10 The transplantation of PDLSCs has been shown to facilitate the regeneration of the cementum, periodontium, and alveolar bone in in vivo experiments conducted on rats, beagles, minipigs, and humans.9 In clinical trials, autologous PDLSC cell membrane sheets were used to treat three patients with periodontitis, and it was found that the formation of neoplastic dentin and periodontium was visible around the periodontal tissue.11 Dental pulp stem cells (DPSCs) can be combined with collagen sponge scaffolds to form complexes for the repair of human mandibular defects12 and the reconstruction of the pulp-dentin complex, thereby generating functional biological tooth roots.13 However, current in vitro organoid constructs lack the requisite functional robustness and physiological relevance, particularly in the context of tooth regeneration, which is inherently challenging because of their high structural complexity. Therefore, it is imperative that a potentially effective tissue engineering approach is developed to regenerate teeth that are functional, biocompatible, sustainable, and reproducible, with the ultimate goal of clinical application.14
In 1942, Waddington initially proposed that the intricate developmental process from genotype to phenotype could be described by the concept of epigenetics. Epigenetic regulation can alter the chromatin state without modifying the DNA sequence, which, in turn, affects gene expression, protein function, and RNA processing.15 Epigenetic modifications can be categorised into three main classifications: These are DNA methylation, histone modifications and chromatin remodelling. DNA methylation is primarily associated with gene regulation and can maintain chromosome integrity. Histone modification is a key process in various biological mechanisms, such as transcriptional regulation, DNA repair, DNA replication, alternative splicing, and chromosome aggregation.16
Epigenetic regulation is implicated in a range of biological processes such as embryonic development,17 disease onset and progression,18 bone homeostasis,19 and stem cell fate determination.20 A reduction in MSC functionality has been identified as a significant underlying cause of numerous pathological conditions. Epigenetic regulation is of critical importance for the establishment and maintenance of the homeostasis equilibrium of MSCs within the body. Previous studies have demonstrated that osteo-/odontogenic differentiation of DMSCs is regulated by epigenetic modifications.21,22 Histone modifications affect osteogenic differentiation by regulating the expression of genes associated with this process. For example, the histone acetyltransferase PCAF has been demonstrated to affect the BMP pathway by elevating histone H3K9 acetylation in the promoter regions of BMP2/4, BMPR1B, and Runx2.23 Additionally, histone methylation represents another histone modification that is closely associated with the regulation of osteogenic-related gene expression, either through activation or repression. This paper presents a systematic and comprehensive review of the current research on BM-MSCs and DMSCs in the field of regenerative medicine. We further elucidate the role of epigenetic regulation in tooth regeneration and focus on the role and mechanism of histone methylation in the regulation of osteogenesis-related genes and tooth-derived stem cell functions.
Osteogenic/odontogenic differentiation-related genes
Bone formation primarily depends on osteoblast differentiation, which is strictly controlled by key transcriptional regulators. These regulators are activated in response to specific external signals and signaling pathways that are regulated during development. The transcription factors (TFs) RUNX2 and SP7 (or Osterix, OSX) are essential for the differentiation of mesenchymal precursor cells into the osteoblast lineage.24 Furthermore, the knockout of either RUNX2 or OSX in vivo impedes the normal formation of bone tissue.
Runx2
RUNX2 is a member of the RUNX family of TFs that forms a heterodimer with core-binding factor b (CBFb). This heterodimer enhances the ability of RUNX2 to bind to DNA and maintains protein stability.25 Runx2 is an indispensable regulator of osteoblast and odontoblast differentiation, controlling the expression of specific genes involved in these processes.
Runx2 has the capacity to exert a variety of functions by regulating several key signaling pathways (such as FGF, Hedgehog, Wnt, and Pthlh) and certain TFs (such as OSX and Dlx5).26 Wnt signaling has been demonstrated to regulate the function of Runx1 and Runx2.27 The canonical Wnt/β-catenin signalling pathway has been shown to regulate osteoblast differentiation. The pathway is initiated by the binding of Wnt to the FZD receptor, which leads to the accumulation of β-catenin. Subsequently, β-catenin moves to the nucleus, leading to the transcription of target genes.27 The noncanonical Wnt signaling ligand, WNT5A, has been identified as a key lineage-specific gene in the regulation of differentiation of dental mesenchymal cells. FGFs are a family of secreted peptides controlling a series of secreted peptides in intrachondral and intramembranous ossification.28 The Twist, Msx2, and PLZF genes are primarily situated upstream of the Runx2 gene. The principal downstream genes were OSX, ATF4, and ZFP521.29 ATF4 has been demonstrated to interact with Runx2, thereby upregulating the expression of osteocalcin (OCN) and Osx.30 BMP signaling is essential for Runx2-dependent induction of the osteoblast phenotype.31 Upon activation of Smads (Smad1/5/8), BMP2 initiates Runx2 gene transcription through its distal P1 promoter and proximal P1 promoter. Runx2 is also phosphorylated by the non-canonical BMP signaling pathway (TAK1-MEK-p38 or ERK), which promotes its association with the co-activator CREB binding protein.32 In addition, BMP also promotes Runx2 acetylation via p300 for Runx2 stabilization.32
Osterix (OSX/Sp7)
OSX/Sp7 is an osteoblast-specific TF expressed in dental embryonic mesenchymal cells. Novel variants of the OSX/Sp7 gene have been identified as a cause of recessive osteogenesis imperfecta, a disorder characterized by bone fragility and hearing impairment.33 Sp7/Osx regulates and influences zebrafish larval tooth development and bone mineralization.34 During osteoblast differentiation, the molecular switch OSX/Sp7 plays a crucial role in the formation of active chromatin states.35 Mice lacking Sp7 are unable to express osteoblast-associated genes such as SPARC(secreted protein, acidic and rich in cysteine), Spp1(secreted phosphoprotein 1)/osteopontin, BSP. Additionally, they exhibited a significant reduction in Cola1 expression, a major component of the bone matrix. Sp7 knockout in mice resulted in serious craniofacial malformations, dentin hypoplasia, and root abnormalities. However, initial tooth morphogenesis and dentin formation in the crowns were not impaired. SP7 is involved in bone and tooth formation through a variety of molecular mechanisms, including FGF and TGF-β/BMP signaling. CHIP-seq data revealed that OSX binds to the BSP promoter, thereby promoting osteoclastic differentiation and mineralization.36 SP7 has been demonstrated to promote dentinogenic cell differentiation by upregulating DSPP, DMP1, nestin, and ALP. Furthermore, Sp7 acts as a cofactor of Dlx5 and physically interacts with Dlx5. The Sp7-Dlx5 complex regulates osteoblast differentiation. Other TFs containing homologous structural domains, including Msx1/2, Satb2, and Alx4, may also interact with Sp7 in the osteogenic genome.34
Dentin salivary phosphoprotein (DSPP)
DSPP represents the primary non-collagenous protein present in dentin, osteoblasts, and alveolar bone, and is essential for the normal mineralization of dentin and enamel.37 The Runx2 and OSX/Sp7 are essential for the differentiation of osteoblasts and odontoblasts, whereas DSPP exerts a regulatory effect on the latter process.38 The functions of DMP1 and DSPP in tooth formation and maintenance can be considered to be complementary and/or synergistic.39 It is hypothesized that Runx2 may affect the development of the tooth embryo before the bell stage, yet not during the subsequent stage. Osx may be consistently expressed in adult dentin and pulp cells, stimulating the differentiation and expression of DSPP via a non-Runx2-dependent signaling pathway in the later phase of tooth development. The BMP2 signaling pathway plays a critical role in the differentiation and maturation of odontoblasts. BMP2-deficient teeth display morphological features similar to dentinogenesis imperfecta (DGI), which is associated with mutations in the DMP1 and DSPP genes. Upregulation of BMP2-driven Dlx3, Dmp1, DSPP, and Sp7 promotes mesenchymal cell differentiation and biomineralization.40
The role of histone methylation regulation in the regulation of osteo-/odontogenic differentiation of DMSCs
Mechanisms of regulation of histone methylation
The basic unit of chromatin is the nucleosome, which is formed by the association of histones (H1, H2A, H2B, H3, and H4) with 147 bp of DNA41,42 (Fig. 1). The DNA is densely wrapped in the nucleus of a eukaryotic cell, forming a tightly organized and compact structure that presents a significant challenge to transcription factors attempting to access the genes. The degree of chromatin openness or accessibility is a crucial factor influencing gene expression. This reflects the transcriptional activity of chromatin and represents a pivotal area of investigation in the regulation of gene expression. This field of study holds considerable significance in the context of epigenetic mapping, cell differentiation and development, and the pathogenesis of various diseases.
Fig. 1
figure 1
Chromatin and nucleosome structure. Nucleosome is the basic structural unit of chromatin, DNA is wrapped around the nucleosome to form chromatin fibers, and then form chromosomes. Nucleosome consists of a dense octamer composed of histones H1, H2A, H2B, H3, and H4, and connecting region DNA
Full size image
Histone modifications within promoter regions are indispensable for regulating chromatin accessibility and gene expression.43 Amino acid residues located at the N-terminus of histones are susceptible to post-translational modifications, including methylation, acetylation, ribosylation, and ubiquitination, among others.44 The pathways involved in these post-translational modifications are clinically approved targets for the treatment of human diseases. Currently, acetylation and methylation have been extensively studied,45 and acetylation of histones enhances gene transcription by loosening histone-DNA complexes to allow transcription factor binding,46 whereas histone methylation can stimulate or inhibit gene transcription.47 Histone methylation has long been implicated in heterochromatin formation and X chromosome inactivation.48,49 Dynamic regulation of histone methylation by site-specific histone methyltransferases (HMTs) and demethylases (HDMs) is essential for establishing and maintaining epigenetic modifications.50 These enzymes are involved in a variety of essential cellular processes such as cell proliferation and differentiation, DNA damage response and senescence, individual development, and cancer. Histone methylation is primarily observed at arginine (S) and lysine (K) residues within the tails of H3/H4. This is mediated by lysine methyltransferases (PKMTs) and arginine methyltransferases (PRMTs).50 Additionally, Lys residues of histones can undergo mono-, dimethyl-, and trimethylation.51 The study of lysine methylation sites in mammalian cells has been a prominent area of research, with six sites–K4, K9, K27, K36, and K79 on H3 and K20 on H4–receiving particular attention.49 Of these, H3K4, H3K36, and H3K79 have been identified as active markers that occupy the active regions of transcribed genes within chromatin. Conversely, H3K9, H3K27, and H4K20 are regarded as repressive markers that are typically related to silencing of gene expression and condensation of chromatin.22 In contrast to H3K9me3, which is a heterochromatin marker, H3K27me3 is usually found in bivalent structural domains together with H3K4me3, and regulates stem cell differentiation. In mammalian cells, H3K27me3 protects heterochromatin tissues following H3K9 methylation deletion.52
The most influential histone methylation sites in dental tissues identified in the current study are H3K4, H3K9, and H3K27. For example, dental follicular progenitor cells and alveolar bone osteoblasts display elevated levels of the H3K4me3 mark, whereas periodontal ligament fibroblasts and odontoblasts exhibit high levels of repressive H3K9me3 and H3K27me3 marks on the OSX, IBSP, and BGLAP/Ocn promoters.47,53 Specific enzymes are responsible for the methylation of H3K4, H3K9, and H3K27, and their activities are essential for this process. H3K4 methyltransferases can be divided into two main categories: SET structural domain-containing proteins (including Mixed-Spectrum Leukemia Proteins, MLLs, and Set1A/B) and unrelated histone methyltransferases (ASH1, SMYD, and PRDM).54 The COMPASS complex, in which mono-, di-, and trimethylates H3K4, comprises MLLs, Set1A/B, WDR5, RbPB5, DYP30, and ASH2. (Fig. 3a) WDR5 is indispensable for the assembly of the SET1 complex and HMT activity. Furthermore, other subunits specifically interact with different KMT2 complexes, thereby increasing the diversity and functional modifications of these methyltransferases. The histone methyltransferases KMT1, KMT2H, and KMT8A/D are involved in methylation at H3K9. The trimethylation of H3K27 is catalyzed by the enzyme EZH254. H3K4 demethylases include the LSD/KDM1 and JARID/KDM5 families. LSD1 removed the methyl group from K4me2 or K4me1, yielding K4me0, whereas KDM5 removed the methyl group from K3me3 or K4me2, producing K4me1.54 The histone demethylase classes JMJD1/KDM3, JMJD2/KDM4, and KDM7 are responsible for H3K9 demethylation, whereas KDM6A/B is involved in H3K27 demethylation (Fig. 2).
Fig. 2
figure 2
KMTs and KDMs mediate histone methylation modifications. a The MLL1/2, MLL3/4 complex catalyzes mono- and dimethylation of H3K4, MLL1 is responsible for H3K4 trimethylation, while KDM5A facilitates the demethylation of H3K4me2 and H3K4me3. LSD1 is responsible for the demethylation of H3K4me1 and H3K4me2. b SETDB1 and G9a are involved in the mono/dimethylation of H3K9, while Suv39h1 is responsible for the di-/trimethylation of H3K9. The demethylation of H3K9 is mediated by the JHDM2/KDM3 family, the JHDM3(JMJD2)/KDM4 family, and PHF8/KDM7B; c EZH2 is the major lysine methyltransferase of H3K27, while KDM6A/B catalyzes the demethylation of H3K27me1/2/3
Full size image
The impact of histone methylation is determined by the maintenance of a precise equilibrium between H3K4me3, H3K9me3, and H3K27me3, which serve as major trimethylation sites. These modifications are essential for the formation of distinct chromatin structures, which, in turn, regulate the expression of downstream genes.47,55,56
Role of H3K4 histone methylation in osteogenic/odontogenic differentiation
MLL family
H3K4 methylation is mediated by the SET1/COMPASS complex, which comprises six catalytic subunits: MLL1-4(KMT2A-D), SETD1A/KMT2F, and SETD1B/KMT2G. The MLL1/2 complex contains the tumor suppressor Menin, which facilitates the recruitment of the MLL1/2 complex to HOX and other sites. The MLL3/4 complex contains NCOA6, PTIP, and PA-1, which contributes to the targeting of the MLL3/4 complex to a distinct set of genes. The MLL3/4 complex also comprises the H3K27 demethylase UTX57 (Fig. 3a).
Fig. 3
figure 3
Role of H3K4 histone methylation modifications in osteo-/odontogenic differentiation of bone/dental-derived MSCs. a The SET1/COMPASS complex mediated H3K4 methylation, is comprised of MLLs, Set1A/B, WDR5, RbPB5, DYP30, and ASH2. b H3K4 histone methylation modifications in osteogenic/odontogenic differentiation MLL1 have been demonstrated to promote Wnt5a transcription and activate downstream transcription factors such as DLX5 and Runx2 by facilitating H3K4me3 generation. This, in turn, affects the transcription of the odontogenic markers DMP1 and DSPP, as well as the genes associated with osteogenic differentiation, OSX, and OCN. Consequently, MLL1 promotes osteogenic differentiation and mineralization of odontogenic stem cells. KDM5A and LSD1 are the major enzymes responsible for demethylating H3K4. Their activity can be influenced by microRNAs, which in turn affects the osteogenic differentiation potential of DMSCs
Full size image
An enhancer is a cis-transcription element critical for the regulation of gene expression. Upon activation, an enhancer binds to various transcription factors, transcriptional co-activators, and RNA Pol II, thereby initiating the activation of target genes. In mammals, H3K4me1 is typically observed in enhancer regions and serves as a hallmark marker of enhancers. Trimethylation of H3K4 (H3K4me3), on the other hand, is predominantly distributed near transcription start sites (TSS) and is associated with transcriptional activation.58 H3K4 monomethylation is dependent on the recruitment of MLL3/4. The MLL1/2 core complex is responsible for the conversion of H3K4 to H3K4me1 and H3K4me2. Methyltransferases MLL3(KMT2C) and MLL4 (KMT2D) are indispensable for the activation of enhancers, cellular differentiation, and overall development. They also regulate the early stages of embryonic development and differentiation of embryonic stem cells,59 and their functions potentially occur independently of their enzymatic activities.60 The MLL2/3-COMPASS complex can bind to the Runx2 P1 promoter, which enriches H3K4me3 levels and decreases H3K4me1 levels. This process has been observed to promote and maintain RUNX2 expression in osteoblasts.61 It has been demonstrated that MLL1 is the enzyme responsible for the deposition of H3K4me3 at the promoters of developmental genes. Conversely, the MLL2-COMPASS-like complex has been demonstrated to deposit H3K4me3 at “bivalent” promoters.60
Previous research has revealed that the catalytic activity of MLL1 is indispensable to skeletal development.60 MLL1 facilitates the deposition of H3K4me3 at the promoter of the Hox gene, a crucial developmental regulator.62 Additionally, it has been observed to regulate dental mesenchymal cell differentiation by modulating the formation of H3K4me3 at the WNT5A promoter.63 MLL1 forms a functional complex with nuclear factor I family member NFIB and targets the developmental genes DLX5 and Cebpa to promote osteogenic differentiation in MSCs.64 SET7 catalyzes H3K4 trimethylation and is significantly expressed in dental papillae at the bell stage, enamel formation stage of developing dental embryos (E17.5, P0 and P3), suggesting that SET7 plays a role in the odontogenic differentiation of dental follicle precursor cells (DFPCs).65 The phytohormone, Ferutinin, has been shown to promote the expression of osteocalcin and collagen 1A1 mRNA and protein levels by activating H3K9 acetylation and H3K4 trimethylation markers in the promoter regions of WNT3A and DVL3 and promote the osteogenic differentiation of DPSCs.66
Histone demethylation is a crucial epigenetic mechanism in the context of tooth-mandibular regeneration.67 The JARID1 family of proteins, LSD1, and the NO66 protein, all of which contain the JmjC structural domain, have been identified as catalysts for H3K4 demethylation. The JARID1 family of proteins demethylates H3K4me2 and H3K4me3, while LSD1 demethylates H3K4me1 and H3K4me2. Additionally, NO66 was identified as a catalyst for all three methylated H3K4 states. These proteins have been observed to exert transcriptional repression through demethylation or recruitment of other inhibitory proteins.
KDM5A
JARID1B/KDM5A has been demonstrated to remove H3K4me2 and H3K4me3, thereby repressing transcription of the Runx2 P1 promoter of osteogenesis-related genes. This results in the inhibition of osteogenic differentiation and exerts a negative regulatory role in bone formation in osteoporotic mice.68,69 KDM5A binds to the promoters of RUNX2, OCN, and OPN, resulting in reduced levels and downregulation of H3K4me3. Several miRNAs have been shown to regulate the function of KDM5A, which in turn regulates osteogenic differentiation. Examples of such miRNAs include miR-10769 and miR-29b-3p.70 During the odontogenic differentiation of human dental pulp cells (hDPCs), there was a notable increase in mRNA expression of KDM5A at the initial stage and in a time-dependent manner throughout the induction of tooth formation. Knockdown of KDM5A resulted in elevated expression of DMP1, DSPP, OSX, and OCN. A notable increase in total H3K4me3 levels was observed, whereas H3K4me2 levels remained unaltered. It has been hypothesized that KDM5A may inhibit the odontogenic differentiation of hDPCs by removing H3K4me3 from the promoters of specific genes. This suggests that KDM5A-dependent histone demethylation is a crucial factor in restorative dentinogenesis.71
LSD1
Lysine-specific demethylase 1 (LSD1, also designated as KDM1A) is a member of the flavin adenine dinucleotide (FAD)-dependent amine oxidase family of demethylases. LSD1 is responsible for removing mono- and bi-methylation modifications of histones H3K4 and H3K9.72 As an initial member of the histone demethylation family, it plays a crucial role in the differentiation of stem cells. LSD1 occupies the binding site for H3K4me2/3, and its activity is essential for the normal formation of bone tissue.73 The administration of LSD1 inhibitors have been demonstrated to prevent osteoporosis after ovarietomy (OVX) in mice by increasing the number of osteoblasts.74 KDM1A/LSD1 has been shown to exhibit dynamic bidirectional effects during osteogenic/odontogenic differentiation of DMSCs. Knockdown of KDM1A increased the expression of BSP, DSPP, DMP1, OSX, Runx2, and DLX in SCAP. Conversely, in an in vivo transplantation experiment, more mineralized tissue formation was observed in the KDM1Ash group than in the control group, accompanied by upregulated expression of BSP and DSPP. Further studies demonstrated that KDM1A can form a protein complex with PLOD2, an important gene involved in the osteogenic differentiation of BM-MSCs, and that this complex can inhibit the osteo-/odontogenic differentiation of SCAP.75 HOXC8 gene has been found to controls the differentiation of spinal motor neurons.76 HOXC8 has been seen to binds directly to the KDM1A promoter region, thereby regulating KDM1A. This results in a negative regulation of the osteo-/odontogenic differentiation of SCAP and the expression of Runx2 and Osx.77
The JmjC structural domain-containing NO66 is a histone demethylase that can catalyze the removal of mono-, di-, and trimethyl marks at H3K4, di- and trimethyl marks at H3K36. In mammals, NO66 is important for bone formation during early development and maintenance of adult bone homeostasis. During osteogenesis induction, NO66 has been observed to interacts with OSX, thereby inhibiting its transcriptional activity and suppressing osteogenesis. This is achieved by removing labeling from the promoters H3K4me3 and H3K36me3 of OSX target genes, including bone salivary protein (BSP), which in turn inhibits osteogenesis.35,78 Although NO66 has been shown to inhibit osteoblast differentiation, whether it plays a similar role in the differentiation of DMSCs into odontoblasts remains unanswered.
Inflammatory condition
During the process of inflammation, various inflammatory factors (e.g., TNF-a, IL-1, IL-6) have been observed to affect the epigenetic characteristics of cells, thereby influencing gene expression.79 Moreover, the inflammatory microenvironment has been demonstrated to impact stem cell differentiation and function.
In patients with chronic periodontitis, bacteria can alter histone modifications in periodontal tissue and activate a series of inflammatory and bone metabolism-related signaling pathways.80,81 Lipopolysaccharide (LPS) has been identified as a pivotal periodontal pathogenic factor, capable of activating NF-κB signaling and promoting the release of inflammatory cytokines such as IL-6 and IL-8. Research has demonstrated that rat dental capsule stem cells possess the capacity to modulate the expression of ERK1/2 and NF-κB signaling pathway via the paracrine pathway. This modulation involves the repression of IL-1β, IL-6, and TNF-α gene expression, while concurrently promoting the expression of IL-4 and TGF-β. This regulatory mechanism contributes to the preservation of dental pulp regeneration in rats afflicted with inflammation.82 In response to LPS stimulation, there was an increase in H3K27me3 enrichment in the extracellular matrix and osteogenic gene promoters, whereas H3K4me3 enrichment increased in the inflammatory response gene promoters.83 In an LPS-induced periodontitis model, the histone lysine methyltransferase SETD1 promoted the expression of inflammatory genes (such as IL-1β, IL-6, and MMP2) in PDLSCs through H3K4 trimethylation.80 SETD1 and p65 have been shown to have a synergistic role in inflammatory regulation. Additionally, LPS increased the intranuclear localization of SETD1, which, in turn, mediated the nuclear translocation of p65 through SETD1 in a p65-dependent manner. Furthermore, the use of an NF-κB inhibitor has been shown to reduce SETD1B expression and enhance osteogenesis in vivo. The expression of KDM5A and LSD1 was increased in LPS-treated hPDLSCs.84 KDM5A knockdown enhanced ALP activity, promoted mineralization, and upregulated the expression of Runx2, OCN, and OPN. ChIP analysis confirmed that KDM5A binds to the miR-495-3p promoter and inhibits the expression of miR-495-3p by demethylating H3K4me3, thereby enhancing HOXC8 transcription and inhibiting the osteogenic differentiation, proliferation, and migration of hPDLSCs in patients with periodontitis.85 miR-590-3p targets LSD1 transcription, upregulates H3K4me2 methylation, and promotes OSX transcription, which in turn facilitates the osteogenic differentiation of hPDLSCs in periodontitis.84
Markers of H3K4me3 activity are present in the promoters of TFs of the early osteogenic lineage, including RUNX2, MSX2, and DLX5. The regulation of histone demethylases/methylases may affect the level of H3K4me2/3 at the promoters of osteo-/odontogenic genes, which in turn affect the transcription of DMP1, DSPP, OSX, and OCN, thereby affecting the osteo-/odontogenic differentiation of DMSCs. KDM5A and LSD1 are the major demethylases of H3K4 (Table 1). Modification of these enzymes or the utilization of miRNAs to affect their transcription may represent promising targets for enhancing dental-derived stem cell-mediated tissue regeneration (Fig. 3b).
Table 1 Role of H3K4 histone methylation modifications in Osteo-/Odontogenic differentiation of bone/dental-derived MSCs
Full size table
Role of H3K9 histone methylation in osteogenic/odontogenic differentiation
Notably, more than 50% of vertebrate genomes are packaged as condensed and transcriptionally repressed heterochromatin.86 Epigenetic markers of heterochromatin are H3K9me2 or H3K9me3. H3K9 methylation is a key epigenetic mark, and this modification has been associated with tissue-specific gene silencing. Theoretically, H3K9 methylation and chromatin densification inhibit transcription in three ways. First, they restrict the entry of RNA polymerase complexes. Second, they can limit the elongation of the transcript or stability of the mRNA. Third, they can block pathways or functions that activate transcription factors.
G9a, GLP, PRDM2, and SUV39H1
In mammals, H3K9 methyltransferases with different catalytic activities and target genes are involved in various cellular processes. These enzymes include the SUV39H1/2, dimeric G9a-GLP, and PRDM families. Synergistic expression of G9a, GLP, PRDM2, and SUV39H1 during mouse dental embryo development is critical for the regulation of tooth development. The differential distribution of these factors in the mesenchyme may be associated with the epigenetic regulation of signaling molecules engaged in mesenchymal-epithelial interactions. SETDB1 catalyzes the monomethylation of H3K9 in the periplasmic region. Furthermore, evidence suggests that SETDB1 may inhibit osteogenic differentiation of BM-MSCs, regulate osteogenic differentiation through the Setdb1/miR-212-3p/Hmgb1 pathway in hFOB cells.87 G9a may serve as a catalyst for mono/dimethylation of H3K9.88 G9a deficiency may result in delayed expression of Shh and BMP2/4 in dental embryos. In addition, G9a may bind to Runx2 to form a complex, which is recruited to the endogenous Runx2 binding site and is directly involved in regulating tooth development.89 In pulp-derived MSCs, low levels of G9a reduce inhibitory H3K9me2 production and promote osteogenesis.90 SUV39H1/2 catalyzes the dimethylation and trimethylation of H3K9 in constitutive heterochromatin. LIM homobox8 (Lhx8) is a highly conserved transcription factor that regulates dentin development and regeneration through the Wnt and TGF-β pathways.91 Elevated Lhx8 levels facilitate the activation of these two pathways, thereby ensuring the maintenance of mesenchymal development during the initial stages of tooth development. Conversely, the differentiation of odontoblast cells is inhibited by Wnt and TGF-β in the subsequent phase. Through RNA hybridization, Zhou et al. discovered that the co-expression pattern of Lhx8 and Suv39h1 in the mesenchyme was consistent with the dynamic expression profiles of the early epithelial signaling molecule FGF8 and the later mesenchymal signaling molecule BMP2.92 In hDPSCs, Suv39h1 forms a complex with Lhx8, which inhibits the expression of the tooth-forming genes Runx2 and DSPP by controlling the formation of H3K9me2/3. (Fig. 4a) The PRDM family members are also involved in H3K9 methylation. Although Prdm3 and Prdm16 have been demonstrated to have a significant impact on craniofacial development by maintaining the spatiotemporal expression of genes associated with cranial neural crest cell development,93 their involvement in DMSCs differentiation has seldom been investigated and the underlying mechanisms remain unclear.
Fig. 4
figure 4
Role of H3K9 histone methylation modifications in osteo-/odontogenic differentiation of bone/dental-derived MSCs. a The synergistic expression of G9a, GLP, PRDM2, and SUV39H1 regulates the expression of signaling molecules involved in mesenchymal-epithelial interactions and tooth development. The deletion of G9a may result in a delayed expression of Shh, and BMP2/4 in the tooth embryo. The Lhx8/Suv39h1 complexes are reciprocally regulated by epithelial-mesenchymal signaling, thereby maintaining a balance between interstitial differentiation and proliferation through H3K9 methylation. b Setdb1 and G9a have been demonstrated to inhibit osteogenic differentiation of DMSCs. However, KDM4B/D and KDM3B have been shown to promote osteo-/odontogenic differentiation of DMSCs by regulating the expression of specific genes
Full size image
JHDM3(JMJD2)/KDM4
The primary enzymes implicated in the demethylation of H3K9 are members of the JHDM2/KDM3, JHDM3(JMJD2)/KDM4, and PHF8/KDM7B families.94 The JHDM2A-C proteins mediate the demethylation of H3K9me1/2, thereby regulating hormone-dependent transcriptional activation. In addition, JHDM3 catalyzes the demethylation of H3K9me2 and H3K9me3 in vitro. KDM3A and KDM4C have been shown to regulate heterodimerization through transcriptional activation of the lectin components NCAPD2 and NCAPG2. Chromatin reorganization inhibits progression of DNA damage and cell senescence.95 KDM4B was identified as an epigenetic coordinator of β-catenin/Smad1-mediated transcription, facilitating the removal of the repressive marker H3K9me3.96 JMJD2/ KDM4B regulates the expression of H3K9me3 in the Runx2 promoter region. Its deletion inhibits osteogenic differentiation of MSCs isolated from oral bones (OMSCs) and promotes oral and maxillofacial bone senescence.97 JMJD2B/ KDM4B facilitates the osteogenic differentiation of hBM-MSCs via regulating the methylation level of H3K9me2 on the Runx2 promoter.98 DLX homologous proteins are involved in the formation of functional dental epithelial tissues and crown morphogenesis, regulating the differentiation of odontoblast cells.99 KDM4B binds to the DLX5 promoter and removes H3K9me3 from the DLX5 gene promoter, and DLX5 plays a key role in osteogenic differentiation by regulating OSX expression.100 The DLX5 gene is highly expressed in DMSCs. DLX5 and KDM4B exert positive effects on BMP signaling. These two factors appear to regulate each other through a positive feedback mechanism.101 The induction of Osterix expression by BMP-2 is facilitated by Dlx5, yet it functions independently of Runx2. This mechanism promotes the osteo-/odontogenic differentiation potential of SCAP by promoting the expression of ALP, DSPP, DMP1, OPN, and OSX.102 In addition, KDM4B can act in conjunction with c-Jun within the JNK signaling pathway, facilitating its recruitment to the DLX5 promoter region, thereby regulating the NGF-mediated osteogenic differentiation of DMSCs.101 KDM4D enhanced the migratory and chemotactic capabilities of SCAP. Using protein profiling technology, co-binding proteins of KDM4D and ribosomal protein S5 (RPS5) were successfully identified. The KDM4D-RPS5 complex jointly promotes osteo-/odontogenic differentiation potential and migratory capacity of SCAP by increasing the expression of ALP, DSPP, and DMP1. These findings suggest that KDM4D is a promising candidate for dentin tissue.103 KDM3B/JMJD1B is a demethylase that alters chromatin modifications through demethylation of H3K9me2. This demethylation process promotes the osteo-/odontogenic differentiation of SCAP by upregulating the expression of specific genes (such as Runx2, Osx, Ocn, and Dspp)67 (Fig. 4b).
Inflammatory condition
While the epigenetic mechanism of KDMs plays a pivotal role in numerous biological processes, evidence suggests that KDM3C exerts an anti-inflammatory effect on oral bacterial infections in periapical and periodontal tissues through the inhibition of NF-κB signaling and osteoclast (OC) production.104 However, its role in the inflammatory response to oral bacterial infections remains unclear.
H3K9me3 is predominantly associated with transcriptional silencing and occupies the promoter region. KMTs and KDMs determine histone methylation and are associated with osteogenic differentiation. KMTs are involved in the repression of the histone marker H3K9, which includes Setdb1 and G9a, and have been demonstrated to inhibit the osteogenic differentiation of DMSCs. However, KDM4B/D and KDM3B have been shown to promote the osteo-/odontogenic differentiation of DMSCs by regulating the expression of specific genes (Table 2).
Table 2 Role of H3K9 histone methylation modifications in Osteo-/Odontogenic differentiation of bone/dental-derived MSCs
Full size table
Role of H3K27 Histone methylation in Osteo-/Odontogenic differentiation
H3K27me3 is associated with gene silencing and predominantly deposited in CpG-rich promoters. H3K27me3-labeled promoters can still be bound by general transcription factors and by suspended Pol II binding, and are therefore considered parthenogenetic heterochromatin, which exhibits a mechanistic profile distinct from that of H3K9me3.
H3K27 methylation is mediated by the Polycomb Repressive Complex 2 (PRC2), which is composed of four core subunits: EZH2, SUZ12, EED, and RBAP46/48.49,105,106 (Fig. 5a) EZH2, the core component of PRC2, facilitates the addition of a methyl group to histone H3K27, ultimately contributing to the maintenance of gene silencing.107 SUZ12 and EED are essential for the enzymatic activity of EZH2. EZH2 contains a carboxy-terminal catalytic SET structural domain. During embryonic development, EZH2 reduces chromatin accessibility and promotes silencing of HOXA and HOXD cluster genes through H3K27 trimethylation-catalyzed nucleosome compaction.108,109
Fig. 5
figure 5
Role of H3K27 histone methylation modification in osteo-/odontogenic differentiation of bone/dental-derived MSCs. a PRC2 protein complex. b EZH2 has been demonstrated to antagonize the function of KDM6A/B and to co-regulate the level of H3K27me3. c PRC2 core subunit EZH2 has been shown to catalyze H3K27 trimethylation and maintain gene silencing, thereby inhibiting the osteogenic differentiation of DMSCs. The specific demethylase KDM6A has been demonstrated to enhance the osteogenic differentiation potential of DMSCs by removing the H3K27me3 mark at the Runx2 and Ocn transcription start sites. KDM6B is required for osteogenic differentiation, with KDM6B binding to BMP2 and HOX genes and co-regulating downstream OSX, BGLAP, and SPP1 transcription
Full size image
EZH2
Numerous studies have identified that Ezh2 as a key regulator of osteogenic differentiation and skeletal development.110,111 Overexpression of EZH2 facilitates lipogenic differentiation of MSCs.112 Under osteogenic induction conditions, EZH2 expression is downregulated and segregated from the RUNX2 promoter. Chromatin accessibility in osteoblasts may be maintained through the synergistic action of Runx2 and its potential co-regulators, including Sp7 and AP-1, on enhancer modules that regulate osteoblast development.113 The conditional knockdown of Ezh2 in undifferentiated MSCs resulted in defects in skeletal morphology and bone formation.114,115 EZH2 exerts its effects not only by directly regulating signaling modules and specific TFs, but also through targeting of numerous novel genes(including ZBTB16, MX1, and FHL1) that mediate the osteogenic differentiation of MSCs110 As a histone methylation transferase, EZH2 enhances migration and chemotaxis of DMSCs.116 Conversely, evidence indicates that it negatively regulates odontogenic differentiation.117,118,119 EZH2 has been observed to inhibit osteogenic differentiation and mineralization of hDPCs through the β-catenin pathway, while simultaneously promoting their proliferation.117
Long non-coding RNAs (lncRNAs) are defined as transcripts exceeding 200 nucleotides in length, and some lncRNAs interact with PRC2 and direct it to specific sites in chromatin.119 In contrast, EZH2, a catalytic component of PRC2, have been observed to bind to RNA and, via EZH2-mediated trimethylation of H3K27, epigenetically silences KLF transcription, repress the expression of ALP, Osx, and OCN, and stimulate mineralization.120 A recent study has found that the mechanosensitive lncRNA SNHG8 exerts a regulatory effect on EZH2 in PDLSCs, thereby inhibiting osteogenic differentiation.121 Mechanical force has been shown to reduce EZH2 expression, accompanied by a delayed decrease in H3K27me3 expression and a significant decrease in H3K4me3 enrichment within the SNHG8 promoter region and 250 bp upstream of it. The results of ChIP-seq corroborated the mutual regulation of SNHG8 and EZH2 and demonstrated that the reduction in SNHG8 expression after mechanical force stretch was attributed to the diminution in the activation of its promoter. This indicated that the reduction in EZH2 expression was a consequence of decreased promoter activation. This indicates that the reduction of EZH2 is a pivotal epigenetic event in the mechanical response related to the maintenance of superenhancer stability and stem cell function. CARMN is an evolutionarily conserved lncRNA that is specific to smooth muscle cells and promotes the odontogenic differentiation of DPCs by impairing EZH2.122
KDM6A
UTX/KDM6A, UTY/KDM6C, and JMJD3/KDM6B are demethylases that remove methyl groups from H3K27me2 and H3K27me3, thereby activating the gene expression. Lysine-specific demethylase 6A (KDM6A), also known as UTX, is a member of the KDM6 family of H3K27 demethylases, with the gene predominantly located at Xp11.3. KDM6A contains a catalytic JmjC structural domain at the C terminus and six tetrapeptide repeat (TPR) structural domains at the N terminus. The catalytic JmjC structural domain enables KDM6A to demethylate H3K27me2/3. However, it has also been posited that the primary function of KDM6A is to recruit additional chromatin regulators.123 KDM6A is an important tumor suppressor124 that regulates a multitude of stem cell functions, including chondrogenesis, macrophage M2 differentiation, muscle differentiation, and neuronal differentiation. KDM6A has been shown to promote the osteogenic differentiation of PDLSCs by catalyzing H3K27me3 demethylation and enhancing the transcription of ALP, Runx2, and OPN.125 Inhibition of EZH2 may preserve the osteogenic potential of PDLSCs following KDM6A knockdown by regulating H3K27me3.126 The utilization of pharmacological agents that target the epigenetic regulation of the KDM6A gene has been demonstrated to effectively restore the osteogenic potential of cells. However, only a few studies have directly demonstrated alterations in KDM6A gene expression in patients with periodontitis.52 LncRNA HOTAIRM1 has been shown to upregulate KDM6A/B expression and suppress EZH2 in a HIF-1α-dependent manner, thereby reducing the distribution of H3K27me3 of ALP, M-CSF, Wnt-3a, Wnt-5a, Wnt-7a, β-catenin and others. This facilitates the transcription of these genes and promotes osteogenesis in hDFSCs.127
KDM6B
KDM6B, a histone demethylase with a Jmj-C structural domain, can convert H3K27me3 (repressive state) to H3K27me1 (activated state) within the central heterochromatin. Additionally, it has the potential to regulate gene transcription through demethylases in a demethylase-dependent or -independent manner. Human KDM6B, also known as JMJD3, contains a JmjC structural domain and a C-terminal fragment embedded in a GATA-like (GATAL) structural domain. JMJD3 counteracts the enzymatic activity of PRC2, thereby regulating the expression of specific genes. JMJD3 functions as a transcription factor that interacts with coactivators to regulate the transcription of target genes independent of its demethylase activity. Additionally, it facilitates transcriptional elongation and gene expression through its effect on RNA polymerase II (Pol II).128 KDM6B has been the subject of extensive studies in the context of immune diseases, cancer, and tumor development as well as in pluripotent stem cells and fate determination.129,130 The osteogenic differentiation of BM-MSCs is a process in which KDM6B is responsible for the removal of methyl groups from histones situated within the promoters of BMP2, BMP4, and HOXC6-1. This consequently regulates the expression of RUNX2.100 In the osteo-/odontogenic differentiation of DMSCs, KDM6B is recruited to the BMP2 promoter, resulting in the removal of the silencing genetic marker H3K27me3 and subsequent activation of transcription of the downstream odontogenic marker gene OSX/sp7, as well as the extracellular matrix genes BGLAP and OPN. The knockdown of KDM6B in DMSCs resulted in decreased ALP activity and mineralization.131 Postmenopausal patients with osteoporosis exhibit a parallel decline in the levels of these markers and mineralization, indicating a significant reduction in the population of bone cells and osteoblasts, as well as demineralization of both dense and cancellous bone. Although DPSCs primarily differentiate into dentin and BM-MSCs differentiate into bone, both dentin and bone formation share similar mineralized matrix components. The function of KDM6B is inhibited by alcohol, which results in impaired osteo/odontogenic differentiation of DPSCs and a reduction in the expression of several genes associated with mineralization, including BMP2, BMP4, OCN, and OPN. This indicates that excessive alcohol consumption may result in cellular damage associated with aberrant tooth development and osteoporosis.132 During the bell stage of human tooth embryo development, miR-93-5p was identified as a differentially expressed miRNA. It was observed to target KDM6B and regulate the H3K27me3 mark on the BMP2 promoter, thereby controlling odontogenic differentiation and dentin formation of DPSCs.133
Inflammatory condition
EZH2 and H3K27me3 expression was reduced in infected pulp tissues and cells. Inhibition of EZH2 suppressed the mRNA expression of IL-1β, IL-6, and IL-8 and proliferation in inflammation-stimulated hDPCs. Inhibition of EZH2 has been demonstrated to promote hDPCs mineralization through the epigenetic regulation of β-catenin expression and the activation of classical Wnt signaling pathways. LPS significantly upregulates EZH2 and H3K27me3 expression in hPDLSCs during the inflammatory process in periodontal tissues. Inhibition of EZH2 suppresses the LPS-induced upregulation of inflammatory factors (IL-6 and TNF-α) expression, cell proliferation and migration. In addition, the knockdown of EZH2 has been demonstrated to facilitate PDLSCs osteogenic differentiation by inhibiting the TLR4/MyD88/NF-κB signaling pathway.118 In the initial stages of periodontitis, KDM6B is recruited to the promoters of IL-6 and IL-12β, leading to H3K27me3 removal and the activation of target genes that regulate the inflammatory response.134 Insulin-like growth factors (IGFs) and their binding proteins (IGFBP5) have been linked to processes such as cell growth, bone repair, and remodeling. IGFBP5 has been identified as a downstream target gene of KDM6B, which forms a protein complex with the BCL6 co-repressor (BCOR). This complex may negatively regulate IGFBP5 transcription by promoting H3K27 methylation in the IGFBP5 promoter region. KDM6B can facilitate periodontal tissue regeneration by removing H3K27me3 from the IGFBP5 promoter region (Table 3).135,136
Table 3 Role of H3K27 histone methylation modification in Osteo-/Odontogenic differentiation of bone/dental-derived MSCs
Full size table
Trimethylation of H3K27 is a repressive epigenetic marker essential for the expression of genes involved in tooth development. The PRC2 core subunit, EZH2, catalyzes H3K27 trimethylation and maintains gene silencing, thereby inhibiting the osteogenic differentiation of DMSCs. The specific demethylase, KDM6A, enhances the osteogenic differentiation potential of DMSCs by removing the H3K27me3 tag at the Runx2 and Ocn transcription start sites. KDM6B is required for osteogenic differentiation, binds to BMP2 and HOX, and regulates the downstream transcription of OSX, BGLAP, and SPP1. Ezh2 and KDM6A are epigenetic switches that function in concert to regulate the level of H3K27me3 in the promoter region of genes, thereby affecting the expression of regulatory genes associated with adipogenesis and osteogenesis137 (Fig. 5c).
Role of bivalent chromatin in osteo-/odontogenic differentiation of DMSCs
Bivalent chromatin
Bivalent chromatin is an epigenetic state that has been identified as a mechanism for lineage commitment and the regulation of developmental gene expression. It is characterized by the presence of H3K4me3 and H3K27me3. Chromatin immunoprecipitation demonstrated that H3K4me3 and H3K27me3 simultaneously occupy certain promoters in epithelial cells. Bivalent chromatin is indispensable for maintaining stem cell pluripotency. It maintains a balance between promoter accessibility and long-range junctions to regulate the expression of developmental genes, such as the regulation of VEGF-responsive angiogenesis138 and the timing of progenitor cell progression to mature neurons,139 affecting tooth organ development65. These modifications, designated as “bivalent structural domains,” serve to maintain a stable state for the genes in question, which subsequently determines whether they are activated or repressed during the process of differentiation. For example, under osteogenic induction conditions, H3K4me3 levels on the RUNX2 promoter in periodontal progenitor cells increased significantly 7 days after induction and then declined rapidly at 14 days. H3K4me3 and H3K27me3 markers on the OSX promoter exhibited a continued decline during osteogenic induction, and ChIP-seq results showed that the inhibitory histone markers H3K9me3 and H3K27me3 were higher in PDL progenitors. These data suggested that alterations in histone dynamics at the promoter level that repress key mineralizing transcription factors regulate mineralization in DMSCs.
H3K4 and H3K27
The presence of bivalent modifications characterized by H3K4me3 and H3K27me3 may exert a pivotal influence on tooth organogenesis by regulating cellular differentiation during tooth germ development. The expression of SET7 and EZH2, which function as methyltransferases, and KDM5B and JMJD3, which act as demethylases, was consistent with the observed expression of H3K4me3 and H3K27me3.65 Epigenetic regulators that modulate bivalent promoters include histone methyltransferases including Ezh2 and MLL1. Inhibition of Ezh2 has been observed to results in the redistribution of bivalent structural domains in transcriptional regulators related to the WNT and Hedgehog pathways in osteoblasts.140 Wnt5a, a significant member of the Wnt ligand family, is a lineage-specific gene that plays an important role in regulating the differentiation of DMSCs.63 It is markedly expressed in both mouse and human dental papillae and is associated with the regulation of odontogenic differentiation and dentin layer formation during the bell stage. WNT5A is associated with bivalent chromatin marks (H3K4me3/H3K27me3) in the stable state. Disassembly of H3K27me3 is a prerequisite for initiation of Wnt5a transcription. This process is subject to stringent regulation by the JMJD3 and MLL complexes, which ultimately determine cell fate commitment in hDPCs. Despite speculation about the involvement of Wnt signaling in the regulation of mineralization, there is a paucity of knowledge regarding the chromatin state of key promoters of mineralization genes, such as RUNX2 and OSX. Furthermore, the effect of these promoters on lineage commitment and matrix secretion in mineralized tissues remains unclear.47
H3K9 and H3K27
SATB2 is a transcription factor implicated in developmental regulation, chromatin remodeling, and transcriptional regulation. It plays a positive regulatory role in the differentiation of osteoblasts, bone formation, and regeneration of MSCs. Mutations in this gene often result in skeletal and dental hypoplasia.141,142 SATB2 promotes the osteogenic differentiation of PDLSCs, DPSCs, and stem cells isolated from human exfoliated deciduous teeth (SHEDs). JHDM1D/KDM7A is a histone demethylase that specifically removes the dimethylation marks of H3K9 and H3K27 from the promoters of target genes.141,143 SATB2 may regulate odontogenic differentiation of hDPSCs by downregulating DKK1 and activating the Wnt/β-catenin signaling pathway through the inhibition of JHDM1D expression. DKK1 is an important target of KDM6A/KDM7A,125 but the mechanism by which DKK1 regulates JHDM1D remains unclear.
H3K4 and H3K36
It has been demonstrated that inflammation and hypoxic niches exert an influence on MSC-mediated tissue regeneration. For instance, oxidative stress, characterized by abnormally elevated reactive oxygen species (ROS) levels, can induce mitochondrial dysfunction, leading to cell death. This, in turn, can diminish the osteogenic and odontogenic capacity of DPCs and intensify inflammatory states.144,145 Hypoxia inhibited the accumulation of superoxide in MSCs mitochondria, upregulated membrane potential, and internalized into damaged cells through extracellular vesicles, thus affecting metabolic status.146
KDM2A is a major member of the JmjC structural domain-containing histone demethylases, which are involved in the demethylation of H3K36 and H3K4, thereby regulating important biological processes, including chromatin remodeling and cell development. For instance, it is responsible for regulating the differentiation of enamel cells and odontoblasts.147 KDM2A has also been demonstrated to modify non-histone proteins, including β-catenin and NF-κB. This enables the regulation of their stability and transcriptional activity.148 KDM2A and BCOR have been shown to form a complex that inhibits osteogenesis. This is achieved by increasing histone H3K4/36 methylation of the epigenetic osteogenic protein (EREG) promoter. This in turn inhibits EREG transcription and subsequent OSX and DLX5 expression.149
SFRP2 is a classical extracellular inhibitor of Wnt signaling. It can also enhance the secretion of osteo-/odontogenic related factors such as IGFBP5, IGFBP4, MMP1, and cell homing related functional proteins CXCL5, CXCL12, CXCL6 in SCAP through paracrine effects. The demethylation process of SFRP2, catalysed by KDM2A, plays a regulatory role in the osteo-/odentigenic differentiation processes of SCAP.150 Under conditions of inflammation and hypoxia, SFRP2 inhibits NF-κB signal transduction by inhibiting Wnt/β-Catenin pathway and enhances osteo-/odontogenic differentiation of SCAP. In conditions of hypoxia, there is an increase in the expression of KDM2A and HIF-1, while BCOR expression remains unaltered. The transcription of SFRP2 is regulated by the demethylation of H3K36me2 and H3K4me3 on the SFRP2 promoter, which ultimately affects the osteo-/odontogenic differentiation function of SCAPs (Table 4).151
Table 4 Role of bivalent histone modifications in Osteo-/Odontogenic differentiation of bone/dental-derived MSCs
Full size table
Despite the advent of massive parallel DNA sequencing techniques, numerous pivotal inquiries into bivalent promoters have persisted since the discovery of bivalence in pluripotent stem cells. To gain further insights into the unique patterns of H3K4me3 and H3K27me3 repair, it is essential to develop new methods that can accurately quantify subtle changes in multiple cell types. Further investigation may elucidate the potential links between bivalent chromatin and the osteo-/odontogenic differentiation of DMSCs, thereby advancing biomedical research on disease treatment and tissue regeneration (Fig. 6).
Fig. 6
figure 6
Role of bivalent histone modifications in the differentiation of bone/dental-derived mesenchymal stem cells
Full size image
Physiological and pathophysiological relevance of histone methylation regulation in DMSCs to health and disease in vivo
The importance of histone methylation regulation of DMSCs in tooth development
During the development of tooth embryos, bivalent modifications are characterized by the presence of H3K4me3 and H3K27me3. The spatiotemporal expression of SET7, EZH2, KDM5B, and JMJD3 plays an instrumental role in this process.65 EZH2 has been shown to antagonize the activity of the Aridl protein, thereby regulating the process of formation and development of dental root forks in murine models through the action of Cdkn2a, which is a critical cell cycle inhibitor.152 ASH2L plays a significant role in the regulation of the expression of key developmental genes, including Shh and Trp63, by modulating H3K4me3 modification. Deficiencies in ASH2L have been observed to result in abnormal differentiation of tooth epithelium, which can ultimately manifest as defects in enamel development.153
Osteoporosis
The gradual accumulation of epigenetic changes associated with the process of aging can lead to abnormal regulation of gene expression, metabolic instability, stem cell aging and/or failure, and tissue homeostasis imbalances.154 H3K4me3, H3K27me3, and H3K36me3 may have undergone ‘remodelling’ during ageing.155 Young exosomes secreted by SHED (SHED-Exos) regulate histone methylation and suppress NF-κB, thereby reversing the senescence of aged tendon stem/progenitor cells (AT-SC) and preserving their tenogenic capacity.156 α-ketoglutaric acid (α-KG), an intermediate derived from the tricarboxylic acid cycle, has demonstrated therapeutic potential in the treatment of age-related osteoporosis by reducing the accumulation of H3K9me3 and H3K27me3, upregulation of BMP signaling, and Nanog expression to restore MSCs function.157 Furthermore, serine synthesis-derived α-KG is imperative for the function of JMJD3, which catalyzes the removal of H3K27me3 at activated T nuclear factor, cytoplasmic 1 (Nfatc1) gene sites, consequently inducing NFATc1 expression and osteoclast maturation.158
The histone demethylase KDM7A has been shown to exerts a pivotal function in regulating bone homeostasis by modulating the differentiation of osteoblasts and osteoclasts. KDM7A has been observed to upregulate the expression of fibroblast activating protein alpha (FAP) and nuclear factor κB ligand receptor activator (RANKL) in BM-MSCs by removing H3K9me2 and H3K27me2 markers from FAP and RANKL promoters. Inhibition of KDM7A has been observed to result in increased FAP expression and inactivation of canonical Wnt signaling. Concurrently, this inhibition has been shown to promote osteoclast differentiation and bone resorption through enhanced RANKL expression.159
Osteoarthritis
KDM6A has been shown to be involved in the process of chondrogenic differentiation of PDLSCs by demethylation of SOX9, Col2a1, and ACAN. The upregulation of KDM6A, or the application of EZH2 inhibitors, has been observed to have the potential to enhance the process of mesenchymal stem cell-mediated cartilage regeneration in cases of inflammatory tissue destruction, a condition that is exemplified by osteoarthritis.126
In osteoarthritis, a reduced methylation of lysine 79 on histone H3 (H3K79me) has been identified as a protective epigenetic mechanism. The study found that preservation of H3K79 methylation with KDM7A/B inhibitors, such as daminozide, was targeted to protect bone joints in intra-articular therapy in mice.160
KDM3A and G9A are a pair of antagonistic histone modification enzymes. KDM3A has been shown to attenuate the ubiquitination of SOX9 by demethylating lysine (K) 68 residues, thereby enhancing the stability of SOX9. In contrast, G9A has been shown to promote the ubiquitination and subsequent degradation of SOX9 by methylating K68 residues. The highly specific G9A inhibitor BIX-01294 has been shown to significantly induce the cartilage differentiation of DPSCs, providing a theoretical basis for enhancing the clinical application of DPSCs in cartilage tissue engineering therapy.161 Consequently, a combination strategy that targets multiple components of epigenetic mechanisms and utilizes a combination of synthetic interactions or immune blocking is expected to enhance the function of DMSCs.
Potential therapeutic targets related to histone methylation regulation of DMSCs
Vital pulp treatment
As previously stated, conventional dentistry is costly, intrusive, and based on a defunct mechanical understanding of dental diseases. In contrast, contemporary biological dentistry utilizes a cellular approach, focusing on the stimulation of cell activity and the promotion of regeneration.162 The development of epigenetic therapy drugs for pulp capping materials currently includes HDAC inhibitors, which have been shown to reduce inflammation and stimulate restorative dentin formation.162,163
However, there is still more that researchers need to learn about how changes to the building blocks of histone methylation affect the regeneration of pulp dentin complex. EZH2 can be used as a potential regulator of pulpitis and regeneration,32,164 and the development of inhibitors or small molecule agents to interfere with its function may be a viable potential therapeutic strategy.
Orthodontic treatment
It has been demonstrated that the accumulation of advanced glycation end products (AGEs) in the periodontal tissue of type 2 diabetes patients contributes to enhanced bone fragility.16 This has been identified as a significant factor hindering orthodontic tooth movement and compromising the efficacy of orthodontic treatment. The targeting of the AGE/RAGE pathway or the enhancement of KDM6B function has been demonstrated to increase the antioxidant capacity of PDLSCs, inhibit cell senescence, and promote osteogenic differentiation. A combination of epigenetic or Wnt pathway modulators with RAGE blockers has the potential to enhance the efficacy of orthodontic treatment in patients with diabetes.165
Maxillofacial bone defect
The treatment of maxillofacial bone defect resulting from periodontitis, trauma, and tumors is contingent upon the restoration of bone tissue.166 Following the degradation of bioactive microspheres, DP-Ak has been observed to release ions and activate sensory nerve cells, resulting in the secretion of calcitonin gene-related peptide (CGRP). The reduction of H3K27me3 levels, achieved by the inhibition of EZH2 and the enhancement of KDM6A, has been demonstrated to promote bone repair.166 The present study investigates the regulatory role of estrogen in DMSCs osteogenesis via the ERα/KDM6B/BMP2 axis in a rat cranioparietal defect model. This process is facilitated by the recruitment of KDM6B to the BMP2 and HOXC6 promoters, resulting in the removal of the H3K27me3 marker and subsequent activation of its transcription.167
Summary and outlook
The methylation of histone lysine or arginine residues has been demonstrated to be of significant importance to gene regulation, as well as other physiological processes. Aberrant histone methylation, which can be precipitated by gene mutation, translocation, or overexpression, frequently results in the onset of developmental defects or diseases. MLL1/4, PRMT1/5, KDM5C, and KDM6B are essential for neurodevelopment, while EZH1/2, MLL4, KDM6B, and EED are implicated in cardiac development. LSD1, MLL1, EED, and G9a impact the hematopoietic system.123 Genetic mutations in H3K4 methyltransferases are linked to syndromes involving bone and facial deformities, intellectual impairment, and often reduced body size and microcephaly.123 Furthermore, it has been determined that mutations in other histone methylation regulators, such as EZH1 and NSD1, can be a causative factor for overgrowth syndrome. The utilisation of small molecule inhibitors of histone modifying enzymes, which correct abnormal methylation, has the potential to function as both novel therapeutic interventions for these diseases and as chemical probes for epigenetic studies.168 Nonetheless, the identification and advancement of small-molecule inhibitors of KMTs and KDMs remains in its nascent stages. Noteworthy endeavors and notable achievements from both academic and pharmaceutical sectors have only surfaced in recent years. Further experimentation is necessary to substantiate the pivotal function of histone methylation-related enzymes in disease processes and to establish a theoretical framework for enhancing the osteo-/odontogenic differentiation function of DMSCs or for the treatment of other diseases.
DMSCs play a pivotal role in tooth development and maintenance of oral tissue health. The capacity for differentiation and self-renewal is important in organizational engineering and regenerative medicine. The odontogenic differentiation of DMSCs is a crucial step in dentin formation. This process is orchestrated at the molecular level by a complex network of signaling pathways, transcription factors, and post-transcriptional and epigenetic regulatory mechanisms, which together facilitate the coordinated expression of a multitude of genes. The histone methylation status is a significant factor in stem cell differentiation. This paper reviews the role of epigenetic histone methylation modifications in tooth development and the potential mechanisms of histone methylation and demethylation enzymes in the regulation of the osteo-/odontogenic differentiation of DMSCs. Methylation of H3K4 and H3K36 is associated with the activation of transcription, whereas that of H3K9 and H3K27 is associated with the repression of transcription.
Osteogenesis comprises three distinct phases: proliferation, matrix maturation, and mineralization. These phases are regulated by transcription factors RUNX2 and OSX. RUNX2 and OSX are early markers of osteoblast/odontoblast differentiation, whereas OCN functions primarily during late osteogenesis. In MSCs, the expression of these genes is negatively regulated by inhibitory epigenetic markers including H3K4me1, H3K9me3, and H3K27me3. Simultaneous activation of the classical Wnt and BMP signaling pathways by RUNX2 and OSX is indispensable for formation and proliferation of the mature osteoblast phenotype. H3K4me2/3 marks were enriched in the promoter regions of osteo-/odontogenic genes of BM-MSCs and DMSCs. The inhibition of histone demethylases KDM1A and KDM5A expression has been demonstrated to facilitate the osteo-/odontogenic differentiation of DMSCs through the activation of Runx2, Osx, and DSPP gene transcription. However, under inflammatory conditions, the accumulation of H3K4me3 may also affect the expression of inflammatory genes. H3K9 methylation is primarily associated with the repression of gene transcription. KDM4B inhibits maxillofacial bone senescence, enhances the proliferation and migration of SCAP, and activates the expression of the osteogenic gene DLX5 by removing the H3K9me2/3 mark. The PRC2 core subunit EZH2 catalyzes the methylation of H3K27, thereby maintaining gene silencing. During the differentiation process of DMSCs, EZH2 and KDM6A/B function in concert to regulate the level of H3K27me3, thereby controlling the expression of osteo-/odontogenic-related genes, including BMP2, OSX, OCN, DSPP, and DMP1.
Bivalent histones may play a significant role in dental organ development and lineage-specific differentiation of MSCs by functioning as regulators of cell differentiation. Co-localization of H3K4me3 and H3K27me3 marks within the promoter region has been observed to alter the expression of certain genes. In pluripotent stem cells, the majority of H3K27me3 peaks were localized to H3K4me3-tagged promoters, indicating that genes with bivalent structural domains are the prevailing phenomenon rather than an exception. This phenomenon also applies to osteogenic differentiation of DMSCs. The inhibitory epigenetic markers H3K9me3 and H3K27me3 exhibit a pattern of bivalent modifications in dental mesenchymal progenitor cells and are predominantly located on OSX during odontogenic differentiation. It has been hypothesized that the histone methylases EZH2, MLL1, and MLL4, in conjunction with the histone demethylases KDM6A/B and KDM7A, may coordinately control the levels of H3K4me3, H3K9me3, and H3K27me3 in specific gene promoters, thereby controlling downstream gene expression. Although most studies have been conducted in vitro, further in vivo studies and animal disease models are necessary to evaluate their potential applications in disease treatment and regenerative medicine. In addition to histone modifications, a number of other epigenetic modifications, including those of RNA and molecular chaperones, contribute to a range of biological processes, including embryonic development, cell differentiation, and maintenance of pluripotency. In conclusion, dynamic modulation of histone methylation plays a significant and multifaceted role in the regulation of chromatin state. Modulating histone functions to control the differentiation of DMSCs is a promising approach for achieving tooth regeneration.
References
Ying, J. et al. Extracellular vesicles in bone and tooth: a state-of-art paradigm in skeletal regeneration. J. Cell Physiol. 234, 14838–14851 (2019).
Google Scholar
Ravi, V. et al. Advances in tooth agenesis and tooth regeneration. Regen. Ther. 22, 160–168 (2023).
CASPubMedPubMed CentralGoogle Scholar
Dominici, M. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315–317 (2006).
CASPubMedGoogle Scholar
Viswanathan, S. et al. Mesenchymal stem versus stromal cells: international Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 21, 1019–1024 (2019).
CASPubMedGoogle Scholar
Chahal, J. et al. Bone marrow mesenchymal stromal cell treatment in patients with osteoarthritis results in overall improvement in pain and symptoms and reduces synovial inflammation. Stem Cells Transl. Med. 8, 746–757 (2019).
CASPubMedPubMed CentralGoogle Scholar
Dorronsoro, A. et al. Mesenchymal stem cell-derived extracellular vesicles reduce senescence and extend health span in mouse models of aging. Aging Cell 20, e13337 (2021).
CASPubMedPubMed CentralGoogle Scholar
Mattei, V. et al. Regenerative potential of DPSCs and revascularization: direct, paracrine or autocrine effect? J. Stem Cell Rev. 17, 1635–46, (2021).
CASGoogle Scholar
Kumar, A. et al. Secretome proteins regulate comparative osteogenic and adipogenic potential in bone marrow and dental stem cells. Biochimie 155, 129–139 (2018).
CASPubMedGoogle Scholar
Zhao, J. et al. Oral cavity-derived stem cells and preclinical models of jaw-bone defects for bone tissue engineering. Stem Cell Res. Ther. 14, 39 (2023).
CASPubMedPubMed CentralGoogle Scholar
Wang, J. et al. Research progress in cell therapy for oral diseases: focus on cell sources and strategies to optimize cell function. Front. Bioeng. Biotech. 12, 1340728 (2024).
Google Scholar
Feng, F. et al. Utility of PDL progenitors for in vivo tissue regeneration: a report of 3 cases. Oral. Dis. 16, 20–28 (2010).
CASPubMedPubMed CentralGoogle Scholar
d’Aquino, R. et al. Human mandible bone defect repair by the grafting of dental pulp stem/progenitor cells and collagen sponge biocomplexes. Eur. Cell Mater. 18, 75–83 (2009).
PubMedGoogle Scholar
Wei, F. et al. Functional tooth restoration by allogeneic mesenchymal stem cell-based bio-root regeneration in swine. Stem Cells Dev. 22, 1752–1762 (2013).
CASPubMedPubMed CentralGoogle Scholar
Sui, B. et al. Mesenchymal condensation in tooth development and regeneration: a focus on translational aspects of organogenesis. Physiol. Rev. 103, 1899–1964 (2023).
CASPubMedGoogle Scholar
Waddington, C. H. The epigenotype. 1942. Int. J. Epidemiol. 41, 10–13 (2012).
CASPubMedGoogle Scholar
Wang, B. & Vashishth, D. Advanced glycation and glycoxidation end products in bone. Bone 176, 116880 (2023).
CASPubMedPubMed CentralGoogle Scholar
Liu, R. et al. NAT10-mediated N4-acetylcytidine mRNA modification regulates self-renewal in human embryonic stem cells. Nucleic Acids Res. 51, 8514–8531 (2023).
CASPubMedPubMed CentralGoogle Scholar
Ling, C. & Rönn, T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 29, 1028–1044 (2019).
CASPubMedPubMed CentralGoogle Scholar
Tesfaye, R. et al. In vitrotracking targets of dynamic super-enhancers to better characterize osteoclastogenesis and to evaluate the effect of diuron on the maturation of human bone cells. Environ. Health Perspect. 131, 67007 (2023).
CASPubMedGoogle Scholar
Sun, Z. et al. Chromatin regulation of transcriptional enhancers and cell fate by the Sotos syndrome gene NSD1. Mol. Cell 83, 2398–2416 (2023).
CASPubMedPubMed CentralGoogle Scholar
Chen, Y. et al. Epigenetic regulation of dental-derived stem cells and their application in pulp and periodontal regeneration. PeerJ. 11, e14550 (2023).
PubMedPubMed CentralGoogle Scholar
Zhang, H. et al. Epigenetic regulation of methylation in determining the fate of dental mesenchymal stem cells. Stem Cells Int. 2022, 5015856 (2022).
PubMedPubMed CentralGoogle Scholar
Li, K. et al. Histone modifications centric-regulation in osteogenic differentiation. Cell Death Discov. 7, 91 (2021).
CASPubMedPubMed CentralGoogle Scholar
Sepulveda, H. et al. Epigenetic signatures at the RUNX2-P1 and Sp7 gene promoters control osteogenic lineage commitment of umbilical cord-derived mesenchymal stem cells. J. Cell Physiol. 232, 2519–2527 (2017).
CASPubMedGoogle Scholar
Komori, T. Regulation of proliferation, differentiation and functions of osteoblasts by Runx2. Int. J. Mol. Sci. 20, 1694 (2019).
CASPubMedPubMed CentralGoogle Scholar
Komori, T. Molecular mechanism of Runx2-dependent bone development. Mol. Cells 43, 168–175 (2020).
CASPubMedPubMed CentralGoogle Scholar
Zhu, S. et al. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov. 10, 71 (2024).
CASPubMedPubMed CentralGoogle Scholar
Ornitz, D. M. & Mari, P. J. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev. 16, 1446–1465 (2002).
CASPubMedGoogle Scholar
Liu, T. M. & Lee, E. H. Transcriptional regulatory cascades in Runx2-dependent bone development. Tissue Eng. Part B Rev. 19, 254–263 (2013).
PubMedGoogle Scholar
Xiao, Y. et al. Advances in the roles of ATF4 in osteoporosis. Biomed. Pharmacother. 169, 115864 (2023).
CASPubMedGoogle Scholar
Phimphilai, M. et al. BMP signaling is required for RUNX2-dependent induction of the osteoblast phenotype. J. Bone Min. Res. 21, 637–646 (2006).
CASGoogle Scholar
Hui, T. et al. EZH2 regulates dental pulp inflammation by direct effect on inflammatory factors. Arch. Oral. Biol. 85, 16–22 (2018).
CASPubMedGoogle Scholar
Fiscaletti, M. et al. Novel variant in Sp7/Osx associated with recessive osteogenesis imperfecta with bone fragility and hearing impairment. Bone 110, 66–75 (2018).
CASPubMedGoogle Scholar
Chen, Z. et al. Sp7/osterix positively regulates and to affect tooth development and bone mineralization in zebrafish larvae. J. Biosci. 44, 127 (2019).
PubMedGoogle Scholar
Sinha, K. et al. Osterix and NO66 histone demethylase control the chromatin of Osterix target genes during osteoblast differentiation. J. Bone Min. Res. 29, 855–865 (2014).
CASGoogle Scholar
Yang, Y. et al. Transcriptional regulation of bone sialoprotein gene expression by Osx. Biochem. Biophys. Res. Commun. 476, 574–579 (2016).
CASPubMedGoogle Scholar
Verdelis, K. et al. Accelerated enamel mineralization in Dspp mutant mice. Matrix Biol. 52-54, 246–259 (2016).
CASPubMedPubMed CentralGoogle Scholar
Chen, S. et al. Runx2, osx, and dspp in tooth development. J. Dent. Res. 88, 904–909 (2009).
CASPubMedPubMed CentralGoogle Scholar
Baba, O. et al. Colocalization of dentin matrix protein 1 and dentin sialoprotein at late stages of rat molar development. Matrix Biol. 23, 371–379 (2004).
CASPubMedGoogle Scholar
Chen, S. et al. Loss of Bmp2 impairs odontogenesis via dysregulating pAkt/pErk/GCN5/Dlx3/Sp7. Res. Sq. 19, rs3299295 (2023).
Google Scholar
Klose, R. & Zhang, Y. Regulation of histone methylation by demethylimination and demethylation. Nat. Rev. Mol. Cell Biol. 8, 307–318 (2007).
CASPubMedGoogle Scholar
Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000).
CASPubMedGoogle Scholar
Lu-Culligan, W. et al. Acetyl-methyllysine marks chromatin at active transcription start sites. Nature 622, 173–179 (2023).
CASPubMedPubMed CentralGoogle Scholar
Tessarz, P. & Kouzarides, T. Histone core modifications regulating nucleosome structure and dynamics. Nat. Rev. Mol. Cell Biol. 15, 703–8, (2014).
CASPubMedGoogle Scholar
Rinaldi, L. & Benitah, S. A. Epigenetic regulation of adult stem cell function. FEBS J. 282, 1589–1604 (2015).
CASPubMedGoogle Scholar
Oishi, T. et al. Contributions of histone tail clipping and acetylation in nucleosome transcription by RNA polymerase II. Nucleic Acid Res. 51, 10364–10374 (2023).
CASPubMedPubMed CentralGoogle Scholar
Gopinathan, G., Luan, X. & Diekwisch, T. G. H. Epigenetic repression of RUNX2 and OSX promoters controls the nonmineralized state of the periodontal ligament. Genes (Basel) 14, 201 (2023).
CASPubMedGoogle Scholar
Peters, A. et al. Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nat. Genet. 30, 77–80 (2002).
CASPubMedGoogle Scholar
Li, Y., Ge, K., Li, T., Cai, R. & Chen, Y. The engagement of histone lysine methyltransferases with nucleosomes: structural basis, regulatory mechanisms, and therapeutic implications. Protein Cell 14, 165–179 (2023).
CASPubMedGoogle Scholar
Liu, C., Lu, F., Cui, X. & Cao, X. Histone methylation in higher plants. Annu. Rev. Plant Biol. 61, 395–420 (2010).
CASPubMedGoogle Scholar
Black, J., Van Rechem, C. & Whetstine, J. R. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol. Cell 48, 491–507 (2012).
CASPubMedGoogle Scholar
Fukuda, K. et al. Epigenetic plasticity safeguards heterochromatin configuration in mammals. Nucleic Acids Res. 51, 6190–6207 (2023).
CASPubMedPubMed CentralGoogle Scholar
Dangaria, S. J., Ito, Y., Luan, X. & Diekwisch, T. G. Differentiation of neural-crest-derived intermediate pluripotent progenitors into committed periodontal populations involves unique molecular signature changes, cohort shifts, and epigenetic modifications. Stem Cells Dev. 20, 39–52 (2011).
CASPubMedGoogle Scholar
Zhang, Q. J. & Liu, Z. P. Histone methylations in heart development, congenital and adult heart diseases. Epigenomics 7, 321–30, (2015).
CASPubMedGoogle Scholar
Li, X. & Li, X. D. Interrogating interactions and modifications of histones in live cells. Cell Chem. Biol. 25, 1–3 (2018).
PubMedGoogle Scholar
Francis, M. et al. Histone methylation: achilles heel and powerful mediator of periodontal homeostasis. J. Dent. Res. 99, 1332–40, (2020).
CASPubMedPubMed CentralGoogle Scholar
Smith, E., Lin, C. & Shilatifard, A. The super elongation complex (SEC) and MLL in development and disease. Genes Dev. 25, 661–672 (2011).
CASPubMedPubMed CentralGoogle Scholar
Li, Y. et al. Structural basis for product specificities of MLL family methyltransferases. Mol. Cell 82, 3810–3825 (2022).
CASPubMedGoogle Scholar
Boileau, R. M., Chen, K. X. & Blelloch, R. Loss of MLL3/4 decouples enhancer H3K4 monomethylation, H3K27 acetylation, and gene activation during embryonic stem cell differentiation. Genome Biol. 24, 41 (2023).
CASPubMedPubMed CentralGoogle Scholar
Cenik, B. K. & Shilatifard, A. COMPASS and SWI/SNF complexes in development and disease. Nat. Rev. Genet. 22, 38–58 (2021).
CASPubMedGoogle Scholar
Rojas, A. et al. Mll-COMPASS complexes mediate H3K4me3 enrichment and transcription of the osteoblast master gene Runx2/p57 in osteoblasts. J. Cell Physiol. 234, 6244–6253 (2019).
CASPubMedGoogle Scholar
Nayak, A., Viale-Bouroncle, S., Morsczeck, C. & Muller, S. The SUMO-specific isopeptidase SENP3 regulates MLL1/MLL2 methyltransferase complexes and controls osteogenic differentiation. Mol. Cell 55, 47–58 (2014).
CASPubMedGoogle Scholar
Zhou, Y. et al. Bivalent histone codes on WNT5A during odontogenic differentiation. J. Dent. Res. 97, 99–107 (2018).
CASPubMedGoogle Scholar
Choi, J. & Lee, H. NFIB-MLL1 complex is required for the stemness and Dlx5-dependent osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. J. Biol. Chem. 299, 105193 (2023).
CASPubMedPubMed CentralGoogle Scholar
Zheng, L. W. et al. Bivalent histone modifications during tooth development. Int. J. Oral. Sci. 6, 205–211 (2014).
CASPubMedPubMed CentralGoogle Scholar
Rolph, D. et al. Ferutinin directs dental pulp-derived stem cells towards the osteogenic lineage by epigenetically regulating canonical Wnt signaling. Biochim. Biophys. Acta Mol. Basis Dis. 1866, 165314 (2020).
CASPubMedGoogle Scholar
Zhang, C. et al. The histone demethylase KDM3B promotes osteo-/odontogenic differentiation, cell proliferation, and migration potential of stem cells from the apical papilla. Stem Cells Int. 2020, 8881021 (2020).
PubMedPubMed CentralGoogle Scholar
Wang, C. et al. KDM5A controls bone morphogenic protein 2-induced osteogenic differentiation of bone mesenchymal stem cells during osteoporosis. Cell Death Dis. 7, e2335 (2016).
CASPubMedPubMed CentralGoogle Scholar
Yan, Y., Yan, H., Qiu, Z. & Long, L. Mechanism of KDM5A-mediated H3K4me3 modification in the osteogenic differentiation of mesenchymal stem cells in steroid-induced osteonecrosis of the femoral head. Int. J. Rheum. Dis. 27, e15090 (2024).
CASPubMedGoogle Scholar
Zhang, X. et al. Extracellular Vesicle-Encapsulated miR-29b-3p released from bone marrow-derived mesenchymal stem cells underpins osteogenic differentiation. Front. Cell Dev. Biol. 8, 581545 (2020).
PubMedGoogle Scholar
Li, Q. M., Li, J. L., Feng, Z. H., Lin, H. C. & Xu, Q. Effect of histone demethylase KDM5A on the odontogenic differentiation of human dental pulp cells. Bioengineered 11, 449–462 (2020).
CASPubMedPubMed CentralGoogle Scholar
Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).
CASPubMedGoogle Scholar
Rummukainen, P. et al. Lysine-Specific Demethylase 1 (LSD1) epigenetically controls osteoblast differentiation. PLoS ONE 17, e0265027 (2022).
CASPubMedPubMed CentralGoogle Scholar
Lv, L. et al. Lysine-specific demethylase 1 inhibitor rescues the osteogenic ability of mesenchymal stem cells under osteoporotic conditions by modulating H3K4 methylation. Bone Res. 4, 16037 (2016).
CASPubMedPubMed CentralGoogle Scholar
Wang, L. et al. KDM1A regulated the osteo/dentinogenic differentiation process of the stem cells of the apical papilla via binding with PLOD2. Cell Prolif. 51, e12459 (2018).
PubMedPubMed CentralGoogle Scholar
Catela, C., Chen, Y., Weng, Y., Wen, K. & Kratsios, P. Control of spinal motor neuron terminal differentiation through sustained Hoxc8 gene activity. Elife 11, e70766 (2022).
CASPubMedPubMed CentralGoogle Scholar
Yang, H., Liang, Y., Cao, Y., Cao, Y. & Fan, Z. Homeobox C8 inhibited the osteo-/dentinogenic differentiation and migration ability of stem cells of the apical papilla via activating KDM1A. J. Cell Physiol. 235, 8432–8445 (2020).
CASPubMedGoogle Scholar
Deng, P., Chen, Q. M., Hong, C. & Wang, C. Y. Histone methyltransferases and demethylases: regulators in balancing osteogenic and adipogenic differentiation of mesenchymal stem cells. Int. J. Oral. Sci. 7, 197–204 (2015).
CASPubMedPubMed CentralGoogle Scholar
Jurdziński, K. T., Potempa, J. & Grabiec, A. M. Epigenetic regulation of inflammation in periodontitis: cellular mechanisms and therapeutic potential. Clin. Epigenetics 12, 186 (2020).
PubMedPubMed CentralGoogle Scholar
Francis, M. et al. SETD1 and NF-kappaB regulate periodontal inflammation through H3K4 trimethylation. J. Dent. Res. 99, 1486–1493 (2020).
CASPubMedPubMed CentralGoogle Scholar
Cao, J. et al. Epigenetic regulation of osteogenic differentiation of periodontal ligament stem cells in periodontitis. Oral. Dis. 29, 2529–2537 (2022).
Google Scholar
Hong, H. et al. Dental follicle stem cells rescue the regenerative capacity of inflamed rat dental pulp through a paracrine pathway. Stem Cell Res. Ther. 11, 333 (2020).
CASPubMedPubMed CentralGoogle Scholar
Liaw, A., Liu, C., Ivanovski, S. & Han, P. The relevance of DNA methylation and histone modification in periodontitis: a scoping review. Cells 11, 3211 (2022).
CASPubMedPubMed CentralGoogle Scholar
Shao, Q., Liu, S., Zou, C. & Ai, Y. Effect of LSD1 on osteogenic differentiation of human periodontal ligament stem cells in periodontitis. Oral. Dis. 29, 1137–1148 (2023).
PubMedGoogle Scholar
Niu, F., Xu, J. & Yan, Y. Histone demethylase KDM5A regulates the functions of human periodontal ligament stem cells during periodontitis via the miR-495-3p/HOXC8 axis. Regen. Ther. 20, 95–106 (2022).
CASPubMedPubMed CentralGoogle Scholar
Padeken, J., Methot, S. & Gasser, S. M. Establishment of H3K9-methylated heterochromatin and its functions in tissue differentiation and maintenance. Nat. Rev. Mol. Cell Biol. 23, 623–640 (2022).
CASPubMedPubMed CentralGoogle Scholar
Zhang, L. et al. Histone methyltransferase Setdb1 mediates osteogenic differentiation by suppressing the expression of miR-212-3p under mechanical unloading. Cell Signal 102, 110554 (2023).
CASPubMedGoogle Scholar
Kamiunten, T. et al. Essential roles of G9a in cell proliferation and differentiation during tooth development. Exp. Cell Res. 357, 202–210 (2017).
CASPubMedGoogle Scholar
Purcell, D. et al. Recruitment of coregulator G9a by Runx2 for selective enhancement or suppression of transcription. J. Cell Biochem. 113, 2406–2414 (2012).
CASPubMedPubMed CentralGoogle Scholar
Shen, W. C. et al. Methylation and PTEN activation in dental pulp mesenchymal stem cells promotes osteogenesis and reduces oncogenesis. Nat. Commun. 10, 2226 (2019).
PubMedPubMed CentralGoogle Scholar
Chen, Z. et al. Lhx8 mediated Wnt and TGFβ pathways in tooth development and regeneration. Biomaterials 63, 35–46 (2015).
PubMedPubMed CentralGoogle Scholar
Zhou, C. et al. FGF8 and BMP2 mediated dynamic regulation of dental mesenchyme proliferation and differentiation via Lhx8/Suv39h1 complex. J. Cell Mol. Med 25, 3051–3062 (2021).
CASPubMedPubMed CentralGoogle Scholar
Shull, L. C. et al. The conserved and divergent roles of Prdm3 and Prdm16 in zebrafish and mouse craniofacial development. Dev. Biol. 461, 132–144 (2020).
CASPubMedPubMed CentralGoogle Scholar
Mosammaparast, N. & Shi, Y. Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu. Rev. Biochem. 79, 155–179 (2010).
CASPubMedGoogle Scholar
Huang, B. et al. KDM3A and KDM4C regulate mesenchymal stromal cell senescence and bone aging via condensin-mediated heterochromatin reorganization. iScience 21, 375–390 (2019).
CASPubMedPubMed CentralGoogle Scholar
Deng, P. et al. Loss of KDM4B exacerbates bone-fat imbalance and mesenchymal stromal cell exhaustion in skeletal aging. Cell Stem Cell 28, 1057–1073 (2021).
CASPubMedPubMed CentralGoogle Scholar
Deng, P. et al. Loss of KDM4B impairs osteogenic differentiation of OMSCs and promotes oral bone aging. Int. J. Oral. Sci. 14, 24 (2022).
CASPubMedPubMed CentralGoogle Scholar
Kang, P. et al. Histone H3K9 demethylase JMJD2B/KDM4B promotes osteogenic differentiation of bone marrow-derived mesenchymal stem cells by regulating H3K9me2 on RUNX2. PeerJ. 10, e13862 (2022).
PubMedPubMed CentralGoogle Scholar
Lézot, F. et al. Physiological implications of DLX homeoproteins in enamel formation. J. Cell Physiol. 216, 688–697 (2008).
PubMedGoogle Scholar
Ye, L. et al. Histone demethylases KDM4B and KDM6B promotes osteogenic differentiation of human MSCs. Cell Stem Cell 11, 50–61 (2012).
CASPubMedPubMed CentralGoogle Scholar
Liu, Z. et al. Epigenetic regulation of NGF-mediated osteogenic differentiation in human dental mesenchymal stem cells. Stem Cells 40, 818–830 (2022).
PubMedPubMed CentralGoogle Scholar
Yang, H. et al. DLX5 and HOXC8 enhance the chondrogenic differentiation potential of stem cells from apical papilla via LINC01013. Stem Cell Res. Ther. 11, 271 (2020).
CASPubMedPubMed CentralGoogle Scholar
Liang, H. et al. KDM4D enhances osteo/dentinogenic differentiation and migration of SCAPs via binding to RPS5. Oral. Dis. 29, 2827–2836 (2022).
Google Scholar
Lee, J. Y. et al. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-kappaB signaling and osteoclastogenesis. FASEB J. 33, 10515–10527 (2019).
CASPubMedPubMed CentralGoogle Scholar
Sparmann, A. & van Lohuizen, M. Polycomb silencers control cell fate, development and cancer. Nat. Rev. Cancer 6, 846–856 (2006).
CASPubMedGoogle Scholar
Wu, Q. et al. Selective translation of epigenetic modifiers affects the temporal pattern and differentiation of neural stem cells. Nat. Commun. 13, 470 (2022).
CASPubMedPubMed CentralGoogle Scholar
Højfeldt, J. et al. Accurate H3K27 methylation can be established de novo by SUZ12-directed PRC2. Nat. Struct. Mol. Biol. 25, 225–232 (2018).
PubMedPubMed CentralGoogle Scholar
Liscovitch-Brauer, N. et al. Profiling the genetic determinants of chromatin accessibility with scalable single-cell CRISPR screens. Nat. Biotechnol. 39, 1270–1277 (2021).
CASPubMedPubMed CentralGoogle Scholar
Margueron, R. et al. Ezh1 and Ezh2 maintain repressive chromatin through different mechanisms. Mol. Cell 32, 503–518 (2008).
CASPubMedPubMed CentralGoogle Scholar
Hemming, S. et al. Identification of novel EZH2 targets regulating osteogenic differentiation in mesenchymal stem cells. Stem Cells Dev. 25, 909–921 (2016).
CASPubMedPubMed CentralGoogle Scholar
Galvan, M. L. et al. Multiple pharmacological inhibitors targeting the epigenetic suppressor enhancer of zeste homolog 2 (Ezh2) accelerate osteoblast differentiation. Bone 150, 115993 (2021).
CASPubMedPubMed CentralGoogle Scholar
Jing, H. et al. Suppression of EZH2 prevents the shift of osteoporotic MSC fate to adipocyte and enhances bone formation during osteoporosis. Mol. Ther. 24, 217–229 (2016).
CASPubMedGoogle Scholar
Hojo, H. et al. Runx2 regulates chromatin accessibility to direct the osteoblast program at neonatal stages. Cell Rep. 40, 111315 (2022).
CASPubMedPubMed CentralGoogle Scholar
Dudakovic, A. et al. Epigenetic control of skeletal development by the histone methyltransferase Ezh2. J. Biol. Chem. 290, 27604–27617 (2015).
CASPubMedPubMed CentralGoogle Scholar
Kobayashi, Y. et al. Ezh2 knockout in mesenchymal cells causes enamel hyper-mineralization. Biochem. Biophys. Res. Commun. 567, 72–78 (2021).
CASPubMedPubMed CentralGoogle Scholar
Ma, H. et al. Enhancer of zeste homolog 2 enhances the migration and chemotaxis of dental mesenchymal stem cells. J. Int. Med. Res. 48, 300060519882149 (2020).
CASPubMedGoogle Scholar
Li, B. et al. EZH2 impairs human dental pulp cell mineralization via the Wnt/beta-Catenin pathway. J. Dent. Res. 97, 571–579 (2018).
CASPubMedGoogle Scholar
Wang, P., Tian, H., Zhang, Z. & Wang, Z. EZH2 regulates lipopolysaccharide-induced periodontal ligament stem cell proliferation and osteogenesis through TLR4/MyD88/NF-kappaB pathway. Stem Cells Int. 2021, 7625134 (2021).
PubMedPubMed CentralGoogle Scholar
Blanco, M. R. & Guttman, M. Re-evaluating the foundations of lncRNA-Polycomb function. EMBO J. 36, 946–966 (2017).
Google Scholar
Li, Z., Guo, X. & Wu, S. Epigenetic silencing of KLF2 by long non-coding RNA SNHG1 inhibits periodontal ligament stem cell osteogenesis differentiation. Stem Cell Res. Ther. 11, 435 (2020).
PubMedPubMed CentralGoogle Scholar
Zhang, Z. et al. Mechanical force-sensitive lncRNA SNHG8 inhibits osteogenic differentiation by regulating EZH2 in hPDLSCs. Cell Signal 93, 110285 (2022).
CASPubMedGoogle Scholar
Li, H. et al. LncRNA CARMN facilitates odontogenic differentiation of dental pulp cells by impairing EZH2. Oral. Dis. 30, 2387–2397 (2023).
PubMedGoogle Scholar
Jambhekar, A., Dhall, A. & Shi, Y. Roles and regulation of histone methylation in animal development. Nat. Rev. Mol. Cell Biol. 20, 625–641 (2019).
CASPubMedPubMed CentralGoogle Scholar
Shi, B. et al. UTX condensation underlies its tumour-suppressive activity. Nature 597, 726–731 (2021).
CASPubMedPubMed CentralGoogle Scholar
Jiang, W., Wang, J. & Zhang, Y. Histone H3K27me3 demethylases KDM6A and KDM6B modulate definitive endoderm differentiation from human ESCs by regulating WNT signaling pathway. Cell Res. 23, 122–130 (2013).
CASPubMedGoogle Scholar
Wang, P. et al. KDM6A promotes chondrogenic differentiation of periodontal ligament stem cells by demethylation of SOX9. Cell Prolif. 51, e12413 (2018).
PubMedGoogle Scholar
Chen, Z. et al. LncRNA HOTAIRM1 promotes dental follicle stem cell-mediated bone regeneration by regulating HIF-1α/KDM6/EZH2/H3K27me3 axis. J. Cell Physiol. 238, 1542–1557 (2023).
CASPubMedGoogle Scholar
Zhang, X., Liu, L., Yuan, X., Wei, Y. & Wei, X. JMJD3 in the regulation of human diseases. Protein Cell 10, 864–882 (2019).
CASPubMedPubMed CentralGoogle Scholar
Xuan, D. et al. Epigenetic modulation in periodontitis: interaction of adiponectin and JMJD3-IRF4 axis in macrophages. J. Cell Physiol. 231, 1090–1096 (2016).
CASPubMedGoogle Scholar
Park, W. Y., Hong, B. J., Lee, J., Choi, C. & Kim, M. Y. H3K27 demethylase JMJD3 employs the NF-κB and BMP signaling pathways to modulate the tumor microenvironment and promote melanoma progression and metastasis. Cancer Res. 76, 161–170 (2016).
CASPubMedGoogle Scholar
Xu, J., Yu, B., Hong, C. & Wang, C. Y. KDM6B epigenetically regulates odontogenic differentiation of dental mesenchymal stem cells. Int. J. Oral. Sci. 5, 200–205 (2013).
CASPubMedPubMed CentralGoogle Scholar
Hoang, M. et al. Alcohol-induced suppression of KDM6B dysregulates the mineralization potential in dental pulp stem cells. Stem Cell Res. 17, 111–121 (2016).
CASPubMedPubMed CentralGoogle Scholar
Wu, S. et al. MicroRNA-93-5p regulates odontogenic differentiation and dentin formation via KDM6B. J. Transl. Med. 22, 54 (2024).
CASPubMedPubMed CentralGoogle Scholar
Wang, P. et al. Jumonji domain-containing protein 3 regulates the early inflammatory response epigenetically in human periodontal ligament cells. Arch. Oral. Biol. 93, 87–94 (2018).
PubMedGoogle Scholar
Liu, D. et al. Demethylation of IGFBP5 by histone demethylase KDM6B promotes mesenchymal stem cell-mediated periodontal tissue regeneration by enhancing osteogenic differentiation and anti-inflammation potentials. Stem Cells 33, 2523–2536 (2015).
CASPubMedGoogle Scholar
Han, N. et al. Local application of IGFBP5 protein enhanced periodontal tissue regeneration via increasing the migration, cell proliferation and osteo/dentinogenic differentiation of mesenchymal stem cells in an inflammatory niche. Stem Cell Res. Ther. 8, 210 (2017).
PubMedPubMed CentralGoogle Scholar
Hemming, S. et al. EZH2 and KDM6A act as an epigenetic switch to regulate mesenchymal stem cell lineage specification. J. Stem Cells 32, 802–815 (2014).
CASGoogle Scholar
Kanki, Y. et al. Bivalent-histone-marked immediate-early gene regulation is vital for VEGF-responsive angiogenesis. Cell Rep. 38, 110332 (2022).
CASPubMedGoogle Scholar
Mätlik, K., Govek, E., Paul, M., Allis, C. & Hatten, M. E. Histone bivalency regulates the timing of cerebellar granule cell development. Genes Dev. 37, 570–589 (2023).
PubMedPubMed CentralGoogle Scholar
Carrasco, M. E., Thaler, R., Nardocci, G., Dudakovic, A. & van Wijnen, A. J. Inhibition of Ezh2 redistributes bivalent domains within transcriptional regulators associated with WNT and Hedgehog pathways in osteoblasts. J. Biol. Chem. 299, 105155 (2023).
CASPubMedPubMed CentralGoogle Scholar
Xin, T. et al. A novel mutation of SATB2 inhibits odontogenesis of human dental pulp stem cells through Wnt/β-catenin signaling pathway. Stem Cell Res. Ther. 12, 595 (2021).
CASPubMedPubMed CentralGoogle Scholar
Cheng, Q. et al. Role of special AT-rich sequence-binding protein 2 in the osteogenesis of human dental mesenchymal stem cells. Stem Cells Dev. 29, 1059–1072 (2020).
CASPubMedGoogle Scholar
Huang, C. et al. Dual-specificity histone demethylase KIAA1718 (KDM7A) regulates neural differentiation through FGF4. Cell Res. 20, 154–165 (2010).
CASPubMedGoogle Scholar
Li, M. et al. A ROS-responsive hydrogel incorporated with dental follicle stem cell-derived small extracellular vesicles promotes dental pulp repair by ameliorating oxidative stress. Bioact. Mater. 36, 524–540 (2024).
CASPubMedPubMed CentralGoogle Scholar
Shirawachi, S. et al. Oxidative stress impairs the calcification ability of human dental pulp cells. BMC Oral. Health 22, 437 (2022).
CASPubMedPubMed CentralGoogle Scholar
Hu, Z. et al. MSCs deliver hypoxia-treated mitochondria reprogramming acinar metabolism to alleviate severe acute pancreatitis injury. Adv. Sci. 10, e2207691 (2023).
Google Scholar
Yi, Q. et al. Spatial and temporal expression of histone demethylase, Kdm2a, during murine molar development. Biotech. Histochem 91, 137–144 (2016).
CASPubMedGoogle Scholar
Martin, M. et al. KDM2A deficiency in the liver promotes abnormal liver function and potential liver damage. Biomolecules 13, 1457 (2023).
CASPubMedPubMed CentralGoogle Scholar
Fan, Z. et al. BCOR regulates mesenchymal stem cell function by epigenetic mechanisms. Nat. Cell Biol. 11, 1002–1009 (2009).
CASPubMedPubMed CentralGoogle Scholar
Yu, G. et al. Demethylation of SFRP2 by histone demethylase KDM2A regulated osteo-/dentinogenic differentiation of stem cells of the apical papilla. Cell Prolif. 49, 330–340 (2016).
CASPubMedPubMed CentralGoogle Scholar
Yang, H. et al. Secreted frizzled-related protein 2 promotes the osteo/odontogenic differentiation and paracrine potentials of stem cells from apical papilla under inflammation and hypoxia conditions. Cell Prolif. 53, e12694 (2020).
PubMedGoogle Scholar
Jing, J. et al. Antagonistic interaction between Ezh2 and Arid1a coordinates root patterning and development via Cdkn2a in mouse molars. Elife 8, 46426 (2019).
Google Scholar
Zhu, X., Ma, Z., Xie, F. & Wang, J. ASH2L, core subunit of H3K4 methylation complex, regulates amelogenesis. J. Dent. Res. 103, 81–90 (2024).
CASPubMedGoogle Scholar
Wang, K. et al. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct. Target Ther. 7, 374 (2022).
CASPubMedPubMed CentralGoogle Scholar
Zhang, W., Qu, J., Liu, G. H. & Belmonte, J. C. I. The ageing epigenome and its rejuvenation. Nat. Rev. Mol. Cell Biol. 21, 137–150 (2020).
CASPubMedGoogle Scholar
Jin, S. et al. Young exosome bio-nanoparticles restore aging-impaired tendon stem/progenitor cell function and reparative capacity. Adv. Mater. 35, e2211602 (2023).
PubMedGoogle Scholar
Wang, Y. et al. Alpha-ketoglutarate ameliorates age-related osteoporosis via regulating histone methylations. Nat. Commun. 11, 5596 (2020).
CASPubMedPubMed CentralGoogle Scholar
Stegen, S., Moermans, K., Stockmans, I., Thienpont, B. & Carmeliet, G. The serine synthesis pathway drives osteoclast differentiation through epigenetic regulation of NFATc1 expression. Nat. Metab. 6, 141–152 (2024).
CASPubMedPubMed CentralGoogle Scholar
Shan, L. et al. Histone demethylase KDM7A regulates bone homeostasis through balancing osteoblast and osteoclast differentiation. Cell Death Dis. 15, 136 (2024).
CASPubMedPubMed CentralGoogle Scholar
Assi, R. et al. Inhibition of KDM7A/B histone demethylases restores H3K79 methylation and protects against osteoarthritis. Ann. Rheuma Dis. 82, 963–973 (2023).
CASGoogle Scholar
Sun, Q. et al. Lysine 68 methylation-dependent SOX9 stability control modulates chondrogenic differentiation in dental pulp stem cells. Adv. Sci. 10, e2206757 (2023).
Google Scholar
Duncan, H. F., Kobayashi, Y., Kearney, M. & Shimizu, E. Epigenetic therapeutics in dental pulp treatment: hopes, challenges and concerns for the development of next-generation biomaterials. Bioact. Mater. 27, 574–593 (2023).
CASPubMedPubMed CentralGoogle Scholar
Duncan, H. F. et al. Release of bio-active dentine extracellular matrix components by histone deacetylase inhibitors (HDACi). Int. Endod. J. 50, 24–38 (2017).
CASPubMedGoogle Scholar
Hui, T. et al. EZH2, a potential regulator of dental pulp inflammation and regeneration. J. Endod. 40, 1132–1138 (2014).
PubMedGoogle Scholar
Ying, Q. et al. AGEs impair osteogenesis in orthodontic force-induced periodontal ligament stem cells through the KDM6B/Wnt self-reinforcing loop. Stem Cell Res. Ther. 15, 431 (2024).
CASPubMedPubMed CentralGoogle Scholar
Tan, B. et al. Biomaterial-based strategies for maxillofacial tumour therapy and bone defect regeneration. Int. J. Oral. Sci. 13, 9 (2021).
CASPubMedPubMed CentralGoogle Scholar
Liu, Z. et al. The ERα/KDM6B regulatory axis modulates osteogenic differentiation in human mesenchymal stem cells. Bone Res. 10, 3 (2022).
CASPubMedPubMed CentralGoogle Scholar
Song, Y., Wu, F. & Wu, J. Targeting histone methylation for cancer therapy: enzymes, inhibitors, biological activity and perspectives. J. Hematol. Oncol. 9, 49 (2016).
PubMedPubMed CentralGoogle Scholar
Download references
Funding
This work was supported by grants from the National Key Research and Development Program (2022YFA1104401), CAMS Innovation Fund for Medical Sciences (2019-I2M-5-031 to Z.P.F.) and grants from Innovation Research Team Project of Beijing Stomatological Hospital, Capital Medical University (NO. CXTD202204 to Z.P.F.).
Author information
Authors and Affiliations
Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
Meijun Hu & Zhipeng Fan
Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
Zhipeng Fan
Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
Zhipeng Fan
Authors
Meijun Hu
View author publications
You can also search for this author inPubMedGoogle Scholar
2. Zhipeng Fan
View author publications
You can also search for this author inPubMedGoogle Scholar
Contributions
Conceptualization, Zhipeng Fan; writing-original draft preparation, Meijun Hu; writing-review and editing, Zhipeng Fan, Meijun Hu. All authors have read and agreed to the published version of the paper.
Corresponding author
Correspondence to Zhipeng Fan.
Ethics declarations
Competing interests
The authors declare no competing interests.
Supplementary information
Plagiarism_Check-2
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
Reprints and permissions
About this article
Check for updates. Verify currency and authenticity via CrossMark
Cite this article
Hu, M., Fan, Z. Role and mechanisms of histone methylation in osteogenic/odontogenic differentiation of dental mesenchymal stem cells. Int J Oral Sci 17, 24 (2025). https://doi.org/10.1038/s41368-025-00353-z
Download citation
Received:26 August 2024
Revised:01 January 2025
Accepted:11 February 2025
Published:26 March 2025
DOI:https://doi.org/10.1038/s41368-025-00353-z
Share this article
Anyone you share the following link with will be able to read this content:
Get shareable link
Sorry, a shareable link is not currently available for this article.
Copy to clipboard
Provided by the Springer Nature SharedIt content-sharing initiative