Back to Journals » Journal of Inflammation Research » Volume 18

Landscape of Histone Posttranslational Modifications in Osteoarthritis

Authors Zhao S, Zheng J, Yue S, Chen X, Dong Y 

Received 27 December 2024

Accepted for publication 30 April 2025

Published 17 June 2025 Volume 2025:18 Pages 7893—7906

DOI https://doi.org/10.2147/JIR.S514599

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Professor Ning Quan



Siyu Zhao, Jia Zheng, Songkai Yue, Xiaoyang Chen, Yonghui Dong

Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China

Correspondence: Yonghui Dong, Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, 450003, People’s Republic of China, Email [email protected]

Abstract: Osteoarthritis (OA) is a complex, progressive, and age-associated disease characterized by aberrant epigenetic expression. Epigenetic analysis has helped clarify the role of histone post-translational modifications (PTMs) in OA. PTMs affect histone structure and function and, therefore, regulate the expression of genes implicated in various biological processes. The roles of histone methylation and acetylation in OA progression—including extracellular collagen degradation and matrix destruction—have been thoroughly analyzed. Though several studies have shown that histone PTMs are related to OA, summative investigations in this area are lacking. The present literature review examines the relationships between histone PTMs and OA. It focuses mainly on methylation, acetylation, phosphorylation, lactylation, ubiquitination, and the roles of the histone methyltransferase (HMT)/histone demethylase (HDMT) and histone acetyltransferase (HAT)/histone deacetylase (HDAC) families in OA development. We used epigenetic tools for discovering new OA treatments. This review offers new perspectives for future studies on OA pathogenesis and treatment.

Keywords: osteoarthritis, histone modifications, epigenetic, cartilage, drug targets

Introduction

Osteoarthritis (OA) is an age-associated disease characterized by chronic joint pain resulting from the degradation of articular cartilage, inflammation of the synovial lining, and changes to the subchondral bone.1 The worldwide incidence of OA in the adult human population is about 303 million. Of these, approximately 96 million are permanently handicapped because of this condition.2 Thus, OA is the leading global cause of age-related disability. As of 2017, there were about 61.2 million reported cases of OA in China.3 In end-stage OA, joint replacement is often required, which entails a significant financial burden and an inherent risk of postoperative complications.

Major risk factors for primary OA include advancing age and obesity.4,5 Recent studies have demonstrated that epigenetic modifications are implicated in OA pathophysiology. The well-structured extracellular matrix (ECM) is the cornerstone of the function of articular cartilage tissue, mainly composed of type II collagen and proteoglycans. The degradation of the cartilage extracellular matrix (ECM) is recognized as a pivotal aspect of OA pathogenesis,6 ultimately contributes to the progressive deterioration and structural collapse of joints. A substantial body of research underscores the crucial role played by four collagenolytic matrix metalloproteinases (MMPs; specifically, MMP-1, 8, 13, and 14) in mediating the breakdown of type II collagen,7 while a distinct class of enzymes known as a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) is primarily responsible for degrading aggrecan.8 Additionally, stromal cells within the synovium and other joint tissues exhibit enhanced production of cytokines and chemokines.9–11 Notably, OA chondrocytes themselves upregulate the expression of pro-catabolic cytokines, such as IL-8, IL-12, and IL-17, which can act in an autocrine or paracrine fashion to exacerbate cartilage degradation, further perpetuating the pathological cascade of OA.12

The results of genome-wide scans and epigenetic studies on joint tissues indicated that the genetic risk polymorphisms associated with OA are mediated through epigenetic mechanisms and their effects on gene expression.12,13 Currently, the emerging epigenetic tools for detecting histone post-translational modifications (PTMs) mainly include the following categories: CUT&Tag, a highly sensitive chromatin analysis technique, combines antibodies with transposases to efficiently capture histone modifications and transcription factor binding sites, making it suitable for small sample applications. High precision mass spectrometry analysis can be employed to identify the types and sites of histone modifications, which is applicable to both quantitative and qualitative research. Antibody array technology, on the other hand, can detect multiple histone modifications using specific antibodies, being suitable for high throughput screening and rapid detection. These tools offer highly sensitive solutions for the detection and functional study of histone modifications, thus propelling the progress in the field of epigenetics. Epigenetic modifications cause reversible and heritable changes in gene function but have no impact on nuclear DNA sequences. The alterations related to epigenetic modifications include DNA methylation, histone modifications, and RNA interference.14 Core histones are evolutionarily conserved and consist of globular domains and flexible charged N-terminal tails. Various enzymes covalently modify the latter mainly at specific lysine and/or arginine residues.15 Histone PTMs affect specific amino acid residues by regulating different enzymes such as writers (add), readers (recognize), or erasers (remove). These markers may control gene expression.16,17 Methylation and acetylation are the most common histone PTMs. The former usually occurs at the lysine (K) residues of histone 3 (H3)3 (Figure 1). Abnormalities in the post-translational regulation of histones are a hallmark of several diseases, including cardiovascular disease (CVD), Alzheimer’s disease and multiple sclerosis (MS). Histone-modifying enzymes are also capable of modifying non-histone proteins. Recent studies (2024) have shown that HDAC3 can drive non-histone deacetylation outside the nucleus. The loss of HDAC3, induced by ECM stiffening, activated Parkin acetylation, thereby stimulating chondrocyte senescence and accelerating the initiation and progression of OA.18 Recently, several studies have shown that OA development and cartilage degeneration are associated with classic histone modifications. Compared to the synovial fibroblasts of healthy individuals, those of patients with OA present with histone hyperacetylation in their interleukin (IL)-6 promoter regions.19 Relative to the chondrocytes of normal controls, those of patients with OA display increased histone H3 at lysine 9 (H3K9) and H3K27 methylation in the SRY-Box Transcription Factor 9 (Sox9) promoter region, leading to decreased expression of Sox9.20 The reduction in Sox9 expression dysregulates chondrocyte metabolism,3 and alters the cartilage phenotype.

Figure 1 Levels of chromatin organization in eukaryotic cells. Eukaryotic chromatin is organized into multiple levels, with nucleosomes as the basic building blocks. A nucleosome consists of 146 bp of DNA wrapped around a core histone octamer, which includes two copies each of H2A, H2B, H3, and H4. The core histones contain globular domains and flexible N-terminal tails, which are the primary sites for post-translational modifications.

Abbreviations: Bp, Base pair; Ac, Acetylation; HMTs, Histone Methyltransferases; HDMTs, Histone Demethylases; HATs, Histone Acetyltransferases; HDACs, Histone Deacetylases.

Despite the high incidence of OA, treatment options have not progressed to the same extent as they have for other musculoskeletal and chronic non-communicable diseases.21 No disease-modifying drugs are currently available for OA treatment. Prescription drugs internationally indicated for OA management merely alleviate pain, and their long-term administration may be associated with side effects and hepatotoxicity.3 Histone PTMs affect a wide range of gene programs. Histone-targeted treatments have demonstrated potential efficacy against OA as they attenuate some of the molecular and physiological changes characteristic of the disease. The present review shows that certain histone modifications regulate OA and may be implicated in its treatment, and an epigenome-based approach could be an efficacious therapeutic modality for OA.

Histones: Their Structure, Functions, and Post-Translational Modifications

Nucleosome surfaces have abundant modifications.22 A recent study showed that covalent histone and DNA modifications alter chromatin organization and function.23 However, the factors affecting nucleosome structure and dynamics, nucleosome-nucleosome interactions, and linker histone-binding and chromatin-binding proteins must be examined to elucidate the functions of chromatin fibers.24

The enzymes transducing histone tail modifications target specific amino acid positions. The N-terminal tails of histone and histone H3 are the most commonly modified. Lee et al suggested that histone tails restrict the access of transcription factors (TF) to DNA. This restriction is removed by directing tail dissociation from DNA, changing the DNA configuration on the histone core, and allowing TF binding.25 Numerous modifications are associated with both active and inactive chromatin regions.

Certain histone amino-terminal modifications synergistically or antagonistically modify the interaction affinities for chromatin-associated proteins, and the latter control the dynamic transitions between transcriptionally active and inactive chromatin states. Methylation, acetylation, and phosphorylation are the most common histone PTMs. Anti-acylated antibodies and ultrasensitive mass spectrometry (MS) have identified and characterized novel histone lysine acylations such as lysine propionylation (Kpr), butyrylation (Kbu), succinylation (Ksucc), crotonylation (Kcr), malonylation (Kma), 2-hydroxyisobutylation (Khib), and lactylation (Kla).26

Histone methylation is vital to the formation of active and inactive genomic regions. Whether it activates or silences transcription depends on the types of residues being modified and the number of methyl groups being added.27 The main classes of HMTs include SET domain lysine methyltransferases, non-SET domain lysine methyltransferases, and arginine methyltransferases.15 There is no pattern to the genomic distribution of histone residue methylation.28 H3K4, H3K36, and H3K79 are methylation markers linked to open chromatin and transcriptional activation and occur in all eukaryotes.29 High degrees of H3K4, H3K79, and H3K36 trimethylation are observed in active gene transcription regions whereas low histone acetylation and high H3K9, H3K27, and H4K20 dimethylation and trimethylation levels are characteristic of the inactive regions.30

Histone acetyltransferases (HATs) mediate histone acetylation, are localized to specific lysine residues on the N-terminal tail of histone, and are associated mainly with transcriptional activity. Active regions exhibit high histone acetylation levels, especially near the promoter and transcription start sites.30 Humans have 18 Zn- or NAD (+)- dependent histone deacetylases (HDACs) that act on acetyl lysine substrates.31 The 18 known HDACs in the human genome are categorized into Classes I–IV depending on their structure, function, localization, and expression patterns. Class I includes HDACs1–3 and 8; Class II includes HDACs4–7 and 9–10; Class III HDACs are also known as sirtuins (Sirt1–7); and Class IV consists of HDAC11 alone.32 Class I HDACs are essential for cell survival and their global deletion is lethal at the embryo stage.32,33 HDACs result in low acetylation levels and heterochromatin and gene silencing. Histone H3 is hyperacetylated in heavily transcribed genome regions but hypoacetylated in silent regions such as telomeres.23

Zhang et al reported the discovery of histone marker lysine lactylation (Kla). Like histone acetylation, histone lactylation facilitates gene transcription under the described conditions.34

Though numerous histone modifications have been detected, the mechanisms regulating epigenetics remain to be elucidated.

Histone Post-Translational Modifications in OA

Histone Methylation in OA

Methylation was one of the first histone PTMs to be discovered. It was reported by Allfrey et al in 1964.35 Histone methylation is often associated with gene expression. Genetic changes and HMT dysregulation are closely related to the deterioration of articular cartilage. Methylation commonly occurs at the lysine residues of histone 3. In mice, post-traumatic OA induction resulted in the rapid decay of H3K79 methylation in articular cartilage. The maintenance of H3K79 methylation is vital to the preservation of joint health and the prevention of OA onset and progression. A recent study (2023) reported a KDM2/7 subfamily inhibitor enhanced H3K79me and exerted protective effects in cartilage in vivo, thereby mitigating cartilage damage and osteophyte and hypertrophic chondrocyte formation in articular cartilage.36 Taken together, the foregoing findings indicate that maintaining H3K79me is essential for preserving joint health and preventing the onset and progression of OA. DOT1L (Disruptor of Telomeric Silencing 1-Like), the gene encoding the H3K79 histone methyltransferase, plays a protective role in OA pathogenesis, as DOT1L-deficient mice exhibit heightened susceptibility to both posttraumatic and spontaneous OA compared to normal controls.1 The loss of DOT1L activity disrupts chondrocyte homeostasis, primarily through its suppression of the Wnt/β-catenin signaling pathway. This pathway, which is highly conserved, includes downstream effectors such as MMP-3 and MMP-13, both of which are known to drive OA progression, highlighting the pathway’s critical involvement in OA development.37 Furthermore, hypoxia induces DOT1L activation in articular cartilage, enhancing its role in maintaining cartilage homeostasis.38 DOT1L also attenuates Wnt signaling by inhibiting sirtuin-1 (SIRT1), thereby slowing OA progression.39 This mechanism is evidence that interactions occur between histone modifications.

Lawson et al reported that conditional ESET histone methyltransferase knockdown results in ectopic articular chondrocyte hypertrophy, articular cartilage degeneration, and other overt signs of terminal differentiation.40 The histone methyltransferase Enhancer of Zeste 2 Polycomb Repressive Complex 2 Subunit (EZH2) regulates the metabolic activity of chondrocytes in OA. Inactivating EZH2 induces micro-RNA (miR)-128a transcription which, in turn, promotes the loss of autophagy-related (Atg)12, represses chondrocyte autophagy, and accelerates OA progression.41 EZH2 is upregulated in OA cartilage and mediates silencing of the Wnt inhibitor secreted frizzled-related protein (SFRP)-1 in OA chondrocytes. Intra-articular injection of the EZH2 inhibitor EPZ005687 impeded OA progression in mice.42 However, a recent study demonstrated that EZH2 ameliorates OA by activating tumor necrosis factor ligand superfamily member (TNFSF)13B.43 This discrepancy may be attributed to the fact that in the first study, EZH2 was inhibited in the cartilage alone through genetic loss whereas in the second study it was inhibited in the entire joint by intra-articular EPZ005687 injection.1

In 2022, our team discovered that PRMT5 (Protein Arginine Methyltransferase 5), an HMT (Histone Methyltransferase), is specifically upregulated in the cartilage of OA (osteoarthritis) patients. Inhibition of PRMT5 suppresses the expression of cartilage matrix-degrading enzymes by reducing the activation of the MAPK (Mitogen-Activated Protein Kinase) and NF-κB (Nuclear Factor kappa B) signaling pathways, thereby slowing OA progression. Additionally, PRMT5 inhibition delays articular cartilage degeneration in the destabilization of the medial meniscus (DMM) mouse model.44

Chondrocytes express the five demethylases LSD1/KDM1, JMJD1A/JHDM2A/KDM3A, KIAA1718/JHDM1D/KDM7A, PHF8/JHDM1F/KDM7B and PHF2/JHDM1E/KDM7C, which are responsible for the demethylation of specific substrates.27 JMJD3 is also called Kdm6b and its protein expression levels and anabolic markers were significantly lower in human OA than in healthy normal cartilages.45 Kdm6b promotes chondrocyte proliferation and hypertrophy during endochondral bone formation.46–49 LSD1 more strongly immunostains in OA than in healthy cartilage. It downregulates the α1 chain of COLIX50 by interacting with the TF SOX9. In human OA chondrocytes, Sox9 suppression is a key target for OA progression.20 Sox9 inactivation is associated with abnormal cartilage differentiation and the obstruction of cartilage and bone formation in OA. Kim et al observed a significant increase in the trimethylation of H3K9 and H3K27 at the SOX9 promoter in OA chondrocytes, as well as a decrease in the acetylation of H3K9, H3K15, H3K18, H3K23, and H3K27, which led to a reduction in the expression of SOX9 and exacerbated the development of OA. Since the synergistic effects of histone modifications at different sites may not clearly determine which modification is responsible for the decrease in SOX9 expression, further research is still needed.51 Recently, a study has pointed out that the overexpression of H3K27 demethylase UTX in mice will lead to the enrichment of H3K27me3 at the SOX9 promoter, thus exacerbating the symptoms of OA. This indicates that controlling H3K27me3 may be a new strategy in the development of OA.52 The post-translational modifications of histones at the H3K4 site are also involved in the occurrence and development of OA. The methyltransferase Set7/9 promotes the expression of COL2A1 by enhancing the level of H3K4me3.53–55

Imbalances between proinflammatory and anti-inflammatory factor expression levels also contribute to extracellular collagen degradation and matrix destruction. Fahmi et al showed that the histone lysine methyltransferase SET-1A was upregulated in OA cartilage compared to normal cartilage,56 and H3K4 dimethylation and trimethylation at the inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 promoters contribute to IL-1β-induced iNOS and COX-2 expression.

The results of the preceding studies indicate that clarifying the roles of HMT/HDMT in OA may facilitate the development and application of efficacious histone-targeted therapy. Table 1 summarizes the representative histone modifications enzymes involved in osteoarthritis.

Table 1 Representative Histone Modifications Enzymes Involved in Osteoarthritis

Histone Acetylation in OA

Hyperacetylation usually marks transcriptionally active genes.61 Lysine acetylation neutralizes the charges on histone tails, weakens their binding to DNA, and relaxes nucleosomal DNA.62 The role of histone lysine acetylation/deacetylation in the progression of OA has been thoroughly studied, aberrant HAT/HDAT-mediated acetylation/deacetylation modifies gene expression in both OA and normal chondrocytes.

In some contexts, HDACs are more sensitive to environmental or developmental cues than the HATs and can provide a regulatory role in transcription.63 Few studies have explored the functions of histone acetyltransferases (HATs) in OA. Only Zupan et al reported that the upregulation of HAT1 is associated with the formation of high-quality bone tissue and a tightly structured trabecular network. They also found that the expression of HAT1 is significantly reduced in postmenopausal osteoarthritis patients, suggesting that dysregulation of HAT1 may exacerbate OA.57 Differential HDAC expression modes are associated with the maintenance of cartilage homeostasis and the initiation and progression of OA. Whereas the upregulation of certain HDACs disrupts chondrocyte homeostasis and accelerates OA progression, the overexpression of other deacetylases is chondroprotective.32 HDAC1,3 and Sirt1,2,3,6,7 are the principal histone deacetylases in normal chondrocytes whereas HDAC1,2,4,7 are highly expressed in OA cartilage (Figure 2).

Figure 2 Alterations of histone-modifying enzymes in OA cartilage. In OA cartilage tissue, the expression levels of HDAC1, 2, 4, 7, 8, PRMT5, and SET-1A were found to be upregulated. This upregulation promotes the increased expression of IL-1β-induced MMPs, which exacerbates cartilage degradation in OA. Meanwhile, SIRT1 expression is decreased in OA cartilage, and the inhibition of its activity also upregulates the expression of cartilage-degrading enzymes MMP-13 and ADAMTS5, thereby promoting the progression of OA.

Abbreviations: HDAC, Histone deacetylase; PRMT5, Protein arginine methyltransferase 5; SET-1A, SET Domain Containing 1A; SIRT1, Sirtuin 1; MMP-13, Matrix metalloproteinase-13; ADAMTS5, A disintegrin and metalloproteinase with thrombospondin motifs 5.

Class I HDACs have been widely studied and include HDAC1, HDAC2, HDAC3, and HDAC8. Genetic deletion of Class I HDACs devastates endochondral bone formation.64 In human chondrocytes, HDAC1, HDAC2, and HDAC3 repress aggrecan and Col2a1,58 and induce IL-1β-induced MMPs expression and catabolic activity.65,66 HDAC2/8 upregulation is observed in patients with OA, prevents chondrogenic differentiation, and inhibits cartilage matrix synthesis.67 You et al suggest that HDAC4 and HDAC8 could drive cartilage degeneration by activating the MAPK signaling pathway. The MAPK (mitogen-activated protein kinase) pathway plays a pivotal role in OA progression, as its overexpression can stimulate the activation of multiple inflammatory factors, leading to the degradation of chondrocytes and the ECM. Consequently, activation of the MAPK pathway hastens the onset and progression of OA.68

The Class IIa HDACs include HDAC4, HDAC5, HDAC7, and HDAC9 (HDAC7b) while the Class IIb HDACs include HDAC6 and HDAC10:69 Class IIa, which has a large C-terminus, and class IIb, which has two deacetylase domains.70

Class II HDACs play a role in maintaining the properties and maturation of chondrocytes. HDAC4 is highly expressed in OA cartilage, suggesting its involvement in promoting the catabolic activity of chondrocytes associated with OA pathogenesis. The expression level of HDAC4 is significantly elevated in OA cartilage tissue, and it may contribute to the pathogenesis of OA by upregulating the catabolic activity of chondrocytes. Additionally, during endochondral ossification, HDAC4 plays a critical regulatory role in the occurrence of chondrocyte hypertrophy, which is one of the typical pathological features of OA cartilage.59,63 HDAC4 is also an upstream mediator of MAPK and promotes ADAMTS4, ADAMTS5, and COX2 expression in rat articular chondrocytes and stimulates IL-1β.68,71 Study shows that MiRNA-222 is minimally expressed in OA cartilage and downregulates MMP-13 by targeting HDAC4 during OA progression.71 HDAC4 and HDAC5 also inhibit Runx2 by transforming growth factor (TGF)-b.72 In human OA, HDAC7 upregulation may contribute to cartilage degradation by elevating MMP-13 expression.48,73 HDAC7 has been demonstrated to induce cartilage damage and ECM degradation through the overexpression of MMP-3 and MMP-13 which is consistent with the HDAC7 knockdown in vitro leading to suppression of inflammatory induced MMP13 gene expression.1,73 Little is known about the functions of HDAC9.

Shen et al reported that HDAC6 was upregulated in the cartilage of a mouse OA model. They also stated that HDAC6 inhibition with tubastatin A (TSA) mitigated oxidative stress in chondrocytes, downregulated apoptotic proteins, increased chondrocyte survival, and suppressed ECM degradation.74

Shinomura et al identified the enhancer elements E1 and E2 in the gene encoding type II collagen and Ea and Eb in the gene encoding aggrecan. They also demonstrated that distinct epigenetic modifications by HDAC10 and SIRT6 control E1 and E2 enhancer activity and thus regulate the expression of type II collagen and aggrecan.75

Class III HDACs include sirtuins (SIRT)1–7. Of these, SIRT1 is the most extensively studied. Research has demonstrated that SIRT1 protects chondrocytes. It is downregulated in human OA cartilage and its inhibition decreases aggrecan expression in both normal and OA human chondrocytes.60 SIRT1 represses IL-1β- and TNF-α-induced cartilage-degrading enzyme expression by modulating the NF-κB pathway achieve a protective effect on cartilage.76,77 OA severity increases with decreasing SIRT1 expression (n = 38).78 Matsushita et al surgically induced OA in SIRT1-CKO mice. Two weeks after the operation, chondrocyte SIRT1 loss accelerated OA development.79 Upregulated type X collagen, MMP-13, ADAMTS5, and apoptotic markers, and acetylated NF-κB p65 in Sirt1-CKO mice compared with the control mice. Another study showed interactions between histone methylation and acetylation. The histone methyltransferase Set7/9 inhibits the histone deacetylase activity of SIRT1 and causes morphology‐dependent Col2a1 transactivation.54 Hence, SIRT1 protects human cartilage by inducing the genes encoding cartilage extracellular matrix (ECM).63 A study revealed that SIRT2 inhibited oxidative stress and the inflammatory response in diabetic OA.80 Griffin et al showed that Sirt3 deletion impaired chondrogenesis in murine bone marrow stem cells whereas Sirt3 expression promoted OA in high-fat-diet-induced mature chondrocytes.81 A study conducted by Wang et al revealed that the SIRT3 level is decreased in mouse OA cartilage and that SIRT3 knockdown induces mitochondrial dysfunction in chondrocytes.82–84 Wu et al overexpressed SIRT3 with lentivirus and plasmids and discovered that it ameliorated OA by regulating chondrocyte autophagy and apoptosis via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway.85 Dai et al revealed that SIRT4 silencing maintained chondrocyte health, downregulated aggrecan, Col2a1, and antioxidant enzymes, and inhibited the inflammatory response.86 There are comparatively few studies on the association between SIRT5 and OA. Only Zhu et al observed that SIRT5-/- mice presented signs of cartilage degeneration and calcification as early as age 40 weeks.87 SIRT6 deficiency exacerbates chondrocyte senescence and accelerates OA progression88 and it also attenuates synovial inflammation by inhibiting proinflammatory cytokine secretion from macrophages in the synovial membrane. In this manner, it mitigates OA severity.89 Ji et al found that intra-articular adenovirus-SIRT6 injection attenuated the surgical destabilization of medial meniscus-induced OA.88 The foregoing findings indicate that SIRT6 may play various roles in OA development.90 SIRT7 regulates cartilage homeostasis and OA development. Yoshizawa et al found that in ATDC5 mice, SIRT7 attenuation increased glycosaminoglycan-rich ECM deposition and the mRNA expression levels of Col2a1 and aggrecan by suppressing SOX9 transcription.91

Class IV histone deacetylases (HDACs): Only one study has demonstrated that HDAC11 is unable to suppress Runx2-induced gene transcription.92 Its precise function remains unclear.

Other Novel Histone Modifications in OA

Previous studies have primarily focused on histone PTMs such as acetylation and methylation. In contrast, few studies have explored or reported on other histone modifications, such as ubiquitination and SUMOylation, in the context of OA.

Histone phosphorylation mediates transcriptional regulation, DNA damage repair, and chromosome condensation during cell division.93 H2A phosphorylation is implicated in mitotic chromatin condensation and transcriptional activation during the G1/S transition.94 Huang et al demonstrated that Wnt10A induces apoptosis in senescent synovial resident stem cells via Wnt/calcium pathway-mediated HDAC5 phosphorylation in the joints of patients with OA.95 A recent study (2025) has revealed the critical role of GRK5-mediated HDAC6 phosphorylation in OA. The study found that G protein-coupled receptor kinase 5 (GRK5), as a HDAC kinase, promotes the phosphorylation of HDAC6, thereby enhancing its deacetylation activity. The phosphorylation of HDAC6 exacerbates mitochondrial dysfunction and ECM in chondrocytes, further promoting the progression of OA.96

Zhao Yingming et al discovered that lactylation is a post-translational histone modification and identified 28 lactylation sites on the core histones in human and mouse cells.34,97 Ouyang et al found that lactate-induced acidification inhibited both chondrocyte proliferation and ECM expression.98 However, the mechanism by which neutral lactate regulates cartilage metabolism remains unclear. Wu et al performed joint CUT&Tag and RNA sequencing analyses and revealed that H3K18la lactylation stimulated the transcription of COL1A2, cartilage oligomeric matrix protein (COMP), ectonucleotide pyrophosphatase/phosphodiesterase (ENPP)1, and transcription factor 7 like 2 (TCF7L2) and may, therefore, protect and repair the ECM in articular cartilage.99,100 Recently, Professor Yu’s team discovered that exogenous lactate-mediated elevation of H4K12la promotes the transcription of PD-1 in microglia, thereby enhancing repair after spinal cord injury.101 Overall, histone lactylation may be part and parcel of the mechanism underlying OA onset and progression.

More comprehensive studies are needed to clarify and understand the precise functions of histone modifications in OA.

Possible Histone Targeted Therapies for Osteoarthritis

Despite the high global incidence of OA, there are few FDA-approved therapies for it, and no currently available drugs prevent or impede the onset of OA.1 Therefore, therapeutic modalities must be able to target a wide range of deregulated gene programs in the entire joint. Histone modifications impact a wide array of gene programs, facilitating the simultaneous regulation of multiple genes linked to OA, such as ECM-degrading enzymes and inflammatory factors, thereby enabling a more comprehensive intervention in OA progression. The interplay between histone modifications can be cooperative or antagonistic, and by targeting multiple modifications, therapeutic efficacy can be enhanced, overcoming the limitations of single-target approaches. In summary, histone-targeted therapy provides an innovative strategy for OA treatment with broad potential for clinical application (Figure 3).

Figure 3 Drugs downregulating MMP-13 and ADAMTS-5 by targeting various HDACs. Trichostatin A (TSA) induced histone H4 hyperacetylation and prevented IL-1β-induced MMP upregulation in human chondrocyte cultures. Valproic acid and MS-275 inhibited Class I HDACs and prevented IL-1β-induced MMP-13 upregulation. SRT1720 upregulated Sirt1, thereby downregulating MMP-13 and ADAMTS-5. The HDAC6 inhibitor ACY-1215 protected human OA chondrocytes and repressed MMPs. The HDAC inhibitor panobinostat inhibited the IL-1β-induced expression of proinflammatory mediators and ECM-degrading enzymes.

Abbreviations: HDACs, histone deacetylase; Col2a1, collagen α1(II); SIRT1, Sirtuin 1; MMP-13, Matrix metalloproteinase-13; ADAMTS5, A disintegrin and metalloproteinase with thrombospondin motifs 5.

DOT1L is the principal H3K79 methyltransferase and it protects against OA by suppressing Wnt signaling. Cartilage-specific Dot1l-knockout (Dot1lCART-KO) mice spontaneously developed severe OA.39 Thus, modulating DOT1L function may be feasible as a therapeutic approach against OA. However, no currently available pharmacological agents induce or activate DOT1L.1 DOT1L is ubiquitously expressed across diverse cell types. Achieving its cell type specific targeting within chondrocytes represents a challenge.102 The lack of specificity may lead to side effects on non-target tissues, which limits its application in the treatment of OA. Recently, Cherifi et al demonstrated that daminozide, an inhibitor of KDM2/7 histone demethylases, counteracts H3K79me deficiency in cartilage and protects against OA.36

Trichostatin A (TSA) exhibits a time- and dose-dependent hyperacetylation effect on histone H4 in chondrocytes. In vitro experiments have confirmed that it can effectively inhibit the upregulation of MMPs induced by IL-1β in human chondrocytes. Additionally, in an experiment where rats received bilateral knee joint injections of TSA (50 μL each time, with an interval of 4 weeks, for a total of 2 times), it was found that compared with the OA group (induced by monoiodoacetate), the cartilage integrity of the rats in the TSA treatment group was significantly improved.103,104 Nasu et al also demonstrated the efficacy of systemic TSA in downregulating various MMPs.53,105 Systemic TSA delivery for more than eight weeks inhibited OA in a mouse DMM model presumably by repressing MMPs. To date, TSA is the only HDACI that has been tested on animal OA models.59

Lotz et al reported that the promising HDAC inhibitor panobinostat upregulated autophagy genes and proteoglycan (PRG)4 and inhibited the IL-1β-induced expression of inflammatory mediators, MMP-3, and MMP-13 by upregulating Forkhead Box Protein (Fox)O1.59 Culley et al found that inhibiting Class I HDACs with valproic acid and entinostat (MS-275) prevented IL-1β-induced MMP-13 upregulation. The selective HDAC3 inhibitor RGFP966 attenuated inflammation in a rat OA model by impeding the nuclear translocation of NF-κB.106 HDAC4 and HDAC8 may promote IL‑1β‑induced catabolic cartilage degradation via the MAPK signaling pathways. Thus, inhibiting HDAC4, HDAC8, or both is a promising OA prevention and treatment strategy.68 Yin et al reported that the HDAC6 inhibitor ACY-1215 protects human OA chondrocytes and suppresses inflammation in them by inhibiting the signal transducer and activator of transcription (STAT)3 and NF-κB signaling pathways.107 SIRT1 inhibition increases apoptosis in chondrocytes whereas the sirtuin activator resveratrol protects them against it.1 Though there is preliminary evidence that HDAC6 inhibitors and SIRT activators promote bone formation and/or prevent bone loss, further research is required to confirm these findings.32 Research has demonstrated that SIRT1 protects chondrocytes. Feige et al developed SRT1720 which increases SIRT1 activity 8.7-fold,108 and observed that SRT1720 treatment delayed OA progression and downregulated both MMP-13 and ADAMTS-5.109 Recent evidence suggests that HDAC inhibitors could effectively protect articular cartilage against destruction by preventing ECM degradation.

As certain broad-spectrum HDAC inhibitors are toxic, they are unsuitable for the treatment of chronic non-life threatening conditions.110 It is challenging to determine in vivo HDAC inhibitor specificity in drug research. HDAC inhibitors are promising as treatments for cancer, inflammation, and neurological diseases, and could prove effective as OA therapy. Several laboratories are developing new HDAC inhibitors that are highly efficacious while triggering few side effects.31

Concluding Remarks

The present review focused on the role of several common histone PTMs and enzymes in OA onset and progression. Prior research has indicated that histone PTMs are ubiquitous in the initiation and development of OA. They regulate the chromatin state, gene expression, and other biological processes. Unlike direct DNA sequence mutations, changes in histone PTMs are usually reversible. Future studies on the mechanism by which histone modifications influence OA could help improve the prevention and treatment of this disease.

Despite certain progress in the research of histone PTMs in OA, numerous challenges still persist. Firstly, there is a wide variety of histone modifications, including methylation, acetylation, phosphorylation, lactylation, etc. Additionally, there may be synergistic or antagonistic interactions among different modifications (such as the interaction between acetylation and methylation). This complexity makes it extremely difficult for researchers to comprehensively analyze the specific mechanisms of each modification in OA. Secondly, the roles of histone - modifying enzymes (such as HDACs, HATs, etc) in OA have been extensively studied. However, these enzymes often function in multiple tissues and cell types, and targeted therapies may cause systemic side effects. Finally, one of the goals of histone PTMs research is to develop targeted therapeutic strategies. Currently, there are no FDA approved histone PTMs targeted drugs for the treatment of OA. Some HDAC inhibitors (such as TSA) have shown potential in animal models, but their toxicity and specificity issues limit their clinical application. A deeper understanding of the role of histone modifications in OA can provide a theoretical basis for the development of precise treatment strategies.

With the rapid development of high-throughput omics technologies, novel histone modifications (such as succinylation, etc) have been continuously discovered. However, the specific functions of these modifications in OA have not been fully clarified. In the future, by combining mass spectrometry technology, integrated proteomics, and bioinformatics analysis, we will conduct an in-depth investigation into the mechanisms of action of these modifications in OA. Additionally, the research focus can be placed on developing more specific modifiers enzyme inhibitors or activators, so as to reduce systemic side effects and enhance the precision and effectiveness of treatment.

This article reviews the role of histone PTMs in OA and its potential therapeutic strategies. Future research needs to combine multi-omics technologies and single-cell analysis to deeply analyze the dynamic changes of histone modifications and their specific mechanisms in OA, providing a theoretical basis for the development of precise therapeutic strategies.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

The present work was supported by Henan Provincial Natural Science Foundation of China (No. 242300420086), the Youth Project Jointly Constructed by Henan Provincial Health Commission and the Ministry (No. SBGJ202103008), and the Henan Young and Middle-aged Health Science and Technology Innovation Excellent Youth Talent Training Project of China (No. YXKC2021047).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Grandi FC, Bhutani N. Epigenetic therapies for osteoarthritis. Trends Pharmacol Sci. 2020;41(8):557–569. doi:10.1016/j.tips.2020.05.008

2. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392(10159):1789–1858. doi:10.1016/S0140-6736(18)32279-7

3. Tong L, Yu H, Huang X, et al. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022;10(1):60. doi:10.1038/s41413-022-00226-9

4. Martel-Pelletier J, Barr AJ, Cicuttini FM, et al. Osteoarthritis. Nat Rev Dis Primers. 2016;2:16072. doi:10.1038/nrdp.2016.72

5. Gorgoulis V, Adams PD, Alimonti A, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–827. doi:10.1016/j.cell.2019.10.005

6. Lohmander LS, Atley LM, Pietka TA, Eyre DR. The release of crosslinked peptides from type II collagen into human synovial fluid is increased soon after joint injury and in osteoarthritis. Arthritis Rheum. 2003;48(11):3130–3139. doi:10.1002/art.11326

7. Little CB, Fosang AJ. Is cartilage matrix breakdown an appropriate therapeutic target in osteoarthritis--insights from studies of aggrecan and collagen proteolysis? Curr Drug Targets. 2010;11(5):561–575. doi:10.2174/138945010791011956

8. Vincent TL, Alliston T, Kapoor M, Loeser RF, Troeberg L, Little CB. Osteoarthritis pathophysiology: therapeutic target discovery may require a multifaceted approach. Clin Geriatr Med. 2022;38(2):193–219. doi:10.1016/j.cger.2021.11.015

9. Brophy RH, Tycksen ED, Sandell LJ, Rai MF. Changes in transcriptome-wide gene expression of anterior cruciate ligament tears based on time from injury. Am J Sports Med. 2016;44(8):2064–2075. doi:10.1177/0363546516643810

10. Pap T, Dankbar B, Wehmeyer C, Korb-Pap A, Sherwood J. Synovial fibroblasts and articular tissue remodelling: role and mechanisms. Semin Cell Dev Biol. 2020;101:140–145. doi:10.1016/j.semcdb.2019.12.006

11. Coutinho de Almeida R, Ramos YFM, Meulenbelt I. Involvement of epigenetics in osteoarthritis. Best Pract Res Clin Rheumatol. 2017;31(5):634–648. doi:10.1016/j.berh.2018.03.002

12. Sandy JD, Chan DD, Trevino RL, Wimmer MA, Plaas A. Human genome-wide expression analysis reorients the study of inflammatory mediators and biomechanics in osteoarthritis. Osteoarthritis Cartilage. 2015;23(11):1939–1945. doi:10.1016/j.joca.2015.03.027

13. Rice SJ, Beier F, Young DA, Loughlin J. Interplay between genetics and epigenetics in osteoarthritis. Nat Rev Rheumatol. 2020;16(5):268–281. doi:10.1038/s41584-020-0407-3

14. Costantino S, Libby P, Kishore R, Tardif JC, El-Osta A, Paneni F. Epigenetics and precision medicine in cardiovascular patients: from basic concepts to the clinical arena. Eur Heart J. 2018;39(47):4150–4158. doi:10.1093/eurheartj/ehx568

15. Teperino R, Schoonjans K, Auwerx J. Histone methyl transferases and demethylases; can they link metabolism and transcription? Cell Metab. 2010;12(4):321–327. doi:10.1016/j.cmet.2010.09.004

16. Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012;48(4):491–507. doi:10.1016/j.molcel.2012.11.006

17. Torres-Perez JV, Irfan J, Febrianto MR, Di Giovanni S, Nagy I. Histone post-translational modifications as potential therapeutic targets for pain management. Trends Pharmacol Sci. 2021;42(11):897–911. doi:10.1016/j.tips.2021.08.002

18. Fu B, Shen J, Zou X, et al. Matrix stiffening promotes chondrocyte senescence and the osteoarthritis development through downregulating HDAC3. Bone Res. 2024;12(1):32. doi:10.1038/s41413-024-00333-9

19. Yang F, Zhou S, Wang C, et al. Epigenetic modifications of interleukin-6 in synovial fibroblasts from osteoarthritis patients. Sci Rep. 2017;7:43592. doi:10.1038/srep43592

20. Kim KI, Park YS, Im GI. Changes in the epigenetic status of the SOX-9 promoter in human osteoarthritic cartilage. J Bone Miner Res. 2013;28(5):1050–1060. doi:10.1002/jbmr.1843

21. Hunter DJ, March L, Chew M. Osteoarthritis in 2020 and beyond: a lancet commission. Lancet. 2020;396(10264):1711–1712. doi:10.1016/S0140-6736(20)32230-3

22. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128(4):693–705. doi:10.1016/j.cell.2007.02.005

23. Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 2014;15(11):703–708. doi:10.1038/nrm3890

24. Chen P, Li W, Li G. Structures and Functions of Chromatin Fibers. Annu Rev Biophys. 2021;50:95–116. doi:10.1146/annurev-biophys-062920-063639

25. Lee DY, Hayes JJ, Pruss D, Wolffe AP. A positive role for histone acetylation in transcription factor access to nucleosomal DNA. Cell. 1993;72(1):73–84. doi:10.1016/0092-8674(93)90051-Q

26. Li D, Zhang L, He Y, et al. Novel histone post-translational modifications in diabetes and complications of diabetes: the underlying mechanisms and implications. Biomed Pharmacother. 2022;156:113984. doi:10.1016/j.biopha.2022.113984

27. El Mansouri FE, Nebbaki SS, Kapoor M, et al. Lysine-specific demethylase 1-mediated demethylation of histone H3 lysine 9 contributes to interleukin 1beta-induced microsomal prostaglandin E synthase 1 expression in human osteoarthritic chondrocytes. Arthritis Res Ther. 2014;16(3):R113. doi:10.1186/ar4564

28. Michalak EM, Burr ML, Bannister AJ, Dawson MA. The roles of DNA, RNA and histone methylation in ageing and cancer. Nat Rev Mol Cell Biol. 2019;20(10):573–589. doi:10.1038/s41580-019-0143-1

29. Trojer P, Reinberg D. Histone lysine demethylases and their impact on epigenetics. Cell. 2006;125(2):213–217. doi:10.1016/j.cell.2006.04.003

30. Barter MJ, Bui C, Young DA. Epigenetic mechanisms in cartilage and osteoarthritis: DNA methylation, histone modifications and microRNAs. Osteoarthritis Cartilage. 2012;20(5):339–349. doi:10.1016/j.joca.2011.12.012

31. Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol. 2014;6(4):a018713. doi:10.1101/cshperspect.a018713

32. Carpio LR, Westendorf JJ. Histone deacetylases in cartilage homeostasis and osteoarthritis. Curr Rheumatol Rep. 2016;18(8):52. doi:10.1007/s11926-016-0602-z

33. Bhaskara S, Chyla BJ, Amann JM, et al. Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol Cell. 2008;30(1):61–72. doi:10.1016/j.molcel.2008.02.030

34. Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575–580. doi:10.1038/s41586-019-1678-1

35. Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc Natl Acad Sci U S A. 1964;51(5):786–794. doi:10.1073/pnas.51.5.786

36. Assi R, Cherifi C, Cornelis FMF, et al. Inhibition of KDM7A/B histone demethylases restores H3K79 methylation and protects against osteoarthritis. Ann Rheum Dis. 2023;82(7):963–973. doi:10.1136/ard-2022-223789

37. Lories RJ, Corr M, Lane NE. To Wnt or not to Wnt: the bone and joint health dilemma. Nat Rev Rheumatol. 2013;9(6):328–339. doi:10.1038/nrrheum.2013.25

38. De Roover A, Nunez AE, Cornelis FM, et al. Hypoxia induces DOT1L in articular cartilage to protect against osteoarthritis. JCI Insight. 2021;6(24). doi:10.1172/jci.insight.150451

39. Monteagudo S, Cornelis FMF, Aznar-Lopez C, et al. DOT1L safeguards cartilage homeostasis and protects against osteoarthritis. Nat Commun. 2017;8:15889. doi:10.1038/ncomms15889

40. Lawson KA, Teteak CJ, Zou J, et al. Mesenchyme-specific knockout of ESET histone methyltransferase causes ectopic hypertrophy and terminal differentiation of articular chondrocytes. J Biol Chem. 2013;288(45):32119–32125. doi:10.1074/jbc.M113.473827

41. Lian WS, Ko JY, Wu RW, et al. MicroRNA-128a represses chondrocyte autophagy and exacerbates knee osteoarthritis by disrupting Atg12. Cell Death Dis. 2018;9(9):919. doi:10.1038/s41419-018-0994-y

42. Chen L, Wu Y, Wu Y, Wang Y, Sun L, Li F. The inhibition of EZH2 ameliorates osteoarthritis development through the Wnt/beta-catenin pathway. Sci Rep. 2016;6:29176. doi:10.1038/srep29176

43. Du X, Chen Y, Zhang Q, et al. Ezh2 ameliorates osteoarthritis by activating TNFSF13B. J Bone Miner Res. 2020;35(5):956–965. doi:10.1002/jbmr.3952

44. Dong Y, Wang P, Yang Y, et al. PRMT5 inhibition attenuates cartilage degradation by reducing MAPK and NF-kappaB signaling. Arthritis Res Ther. 2020;22(1):201. doi:10.1186/s13075-020-02304-x

45. Dai J, Yu D, Wang Y, et al. Kdm6b regulates cartilage development and homeostasis through anabolic metabolism. Ann Rheum Dis. 2017;76(7):1295–1303. doi:10.1136/annrheumdis-2016-210407

46. Zhang F, Xu L, Xu L, et al. JMJD3 promotes chondrocyte proliferation and hypertrophy during endochondral bone formation in mice. J Mol Cell Biol. 2015;7(1):23–34. doi:10.1093/jmcb/mjv003

47. Jin Y, Liu Z, Li Z, et al. Histone demethylase JMJD3 downregulation protects against aberrant force-induced osteoarthritis through epigenetic control of NR4A1. Int J Oral Sci. 2022;14(1):34. doi:10.1038/s41368-022-00190-4

48. Zhang M, Egan B, Wang J. Epigenetic mechanisms underlying the aberrant catabolic and anabolic activities of osteoarthritic chondrocytes. Int J Biochem Cell Biol. 2015;67:101–109. doi:10.1016/j.biocel.2015.04.019

49. Rodova M, Lu Q, Li Y, et al. Nfat1 regulates adult articular chondrocyte function through its age-dependent expression mediated by epigenetic histone methylation. J Bone Miner Res. 2011;26(8):1974–1986. doi:10.1002/jbmr.397

50. Durand AL, Dufour A, Aubert-Foucher E, et al. The lysine specific demethylase-1 negatively regulates the COL9A1 gene in human articular chondrocytes. Int J Mol Sci. 2020;21(17):6322. doi:10.3390/ijms21176322

51. Ukita M, Matsushita K, Tamura M, Yamaguchi T. Histone H3K9 methylation is involved in temporomandibular joint osteoarthritis. Int J Mol Med. 2020;45(2):607–614. doi:10.3892/ijmm.2019.4446

52. Lian WS, Wu RW, Ko JY, et al. Histone H3K27 demethylase UTX compromises articular chondrocyte anabolism and aggravates osteoarthritic degeneration. Cell Death Dis. 2022;13(6):538. doi:10.1038/s41419-022-04985-5

53. Simon TC, Jeffries MA. The Epigenomic Landscape in Osteoarthritis. Curr Rheumatol Rep. 2017;19(6):30. doi:10.1007/s11926-017-0661-9

54. Oppenheimer H, Kumar A, Meir H, et al. Set7/9 impacts COL2A1 expression through binding and repression of SirT1 histone deacetylation. J Bone Miner Res. 2014;29(2):348–360. doi:10.1002/jbmr.2052

55. Okamoto K, Araki Y, Aizaki Y, Tanaka S, Kadono Y, Mimura T. Regulation of cytokine and chemokine expression by histone lysine methyltransferase MLL1 in rheumatoid arthritis synovial fibroblasts. Sci Rep. 2024;14(1):10610. doi:10.1038/s41598-024-60860-7

56. El Mansouri FE, Chabane N, Zayed N, et al. Contribution of H3K4 methylation by SET-1A to interleukin-1-induced cyclooxygenase 2 and inducible nitric oxide synthase expression in human osteoarthritis chondrocytes. Arthritis Rheum. 2011;63(1):168–179. doi:10.1002/art.27762

57. Vrtacnik P, Zupan J, Mlakar V, et al. Epigenetic enzymes influenced by oxidative stress and hypoxia mimetic in osteoblasts are differentially expressed in patients with osteoporosis and osteoarthritis. Sci Rep. 2018;8(1):16215. doi:10.1038/s41598-018-34255-4

58. Hong S, Derfoul A, Pereira-Mouries L, Hall DJ. A novel domain in histone deacetylase 1 and 2 mediates repression of cartilage-specific genes in human chondrocytes. FASEB J. 2009;23(10):3539–3552. doi:10.1096/fj.09-133215

59. Ohzono H, Hu Y, Nagira K, et al. Targeting FoxO transcription factors with HDAC inhibitors for the treatment of osteoarthritis. Ann Rheum Dis. 2023;82(2):262–271. doi:10.1136/ard-2021-221269

60. Fujita N, Matsushita T, Ishida K, et al. Potential involvement of SIRT1 in the pathogenesis of osteoarthritis through the modulation of chondrocyte gene expressions. J Orthop Res. 2011;29(4):511–515. doi:10.1002/jor.21284

61. Delcuve GP, Khan DH, Davie JR. Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Clin Epigenet. 2012;4(1):5. doi:10.1186/1868-7083-4-5

62. Clayton AL, Hazzalin CA, Mahadevan LC. Enhanced histone acetylation and transcription: a dynamic perspective. Mol Cell. 2006;23(3):289–296. doi:10.1016/j.molcel.2006.06.017

63. Dvir-Ginzberg M, Gagarina V, Lee EJ, Hall DJ. Regulation of cartilage-specific gene expression in human chondrocytes by SirT1 and nicotinamide phosphoribosyltransferase. J Biol Chem. 2008;283(52):36300–36310. doi:10.1074/jbc.M803196200

64. Pillai R, Coverdale LE, Dubey G, Martin CC. Histone deacetylase 1 (HDAC-1) required for the normal formation of craniofacial cartilage and pectoral fins of the zebrafish. Dev Dyn. 2004;231(3):647–654. doi:10.1002/dvdy.20168

65. Culley KL, Hui W, Barter MJ, et al. Class I histone deacetylase inhibition modulates metalloproteinase expression and blocks cytokine-induced cartilage degradation. Arthritis Rheum. 2013;65(7):1822–1830. doi:10.1002/art.37965

66. Meng F, Li Z, Zhang Z, et al. MicroRNA-193b-3p regulates chondrogenesis and chondrocyte metabolism by targeting HDAC3. Theranostics. 2018;8(10):2862–2883. doi:10.7150/thno.23547

67. Mao G, Hu S, Zhang Z, et al. Exosomal miR-95-5p regulates chondrogenesis and cartilage degradation via histone deacetylase 2/8. J Cell Mol Med. 2018;22(11):5354–5366. doi:10.1111/jcmm.13808

68. Wang P, Mao Z, Pan Q, et al. Histone deacetylase-4 and histone deacetylase-8 regulate interleukin-1beta-induced cartilage catabolic degradation through MAPK/JNK and ERK pathways. Int J Mol Med. 2018;41(4):2117–2127. doi:10.3892/ijmm.2018.3410

69. Bradley EW, Carpio LR, van Wijnen AJ, McGee-Lawrence ME, Westendorf JJ. Histone deacetylases in bone development and skeletal disorders. Physiol Rev. 2015;95(4):1359–1381. doi:10.1152/physrev.00004.2015

70. Yoon S, Eom GH. HDAC and HDAC inhibitor: from cancer to cardiovascular diseases. Chonnam Med J. 2016;52(1):1–11. doi:10.4068/cmj.2016.52.1.1

71. Song J, Jin EH, Kim D, Kim KY, Chun CH, Jin EJ. MicroRNA-222 regulates MMP-13 via targeting HDAC-4 during osteoarthritis pathogenesis. BBA Clin. 2015;3:79–89. doi:10.1016/j.bbacli.2014.11.009

72. Kang JS, Alliston T, Delston R, Derynck R. Repression of Runx2 function by TGF-beta through recruitment of class II histone deacetylases by Smad3. EMBO J. 2005;24(14):2543–2555. doi:10.1038/sj.emboj.7600729

73. Higashiyama R, Miyaki S, Yamashita S, et al. Correlation between MMP-13 and HDAC7 expression in human knee osteoarthritis. Mod Rheumatol. 2010;20(1):11–17. doi:10.3109/s10165-009-0224-7

74. Shen Z, Ji K, Cai Z, et al. Inhibition of HDAC6 by Tubastatin A reduces chondrocyte oxidative stress in chondrocytes and ameliorates mouse osteoarthritis by activating autophagy. Aging. 2021;13(7):9820–9837. doi:10.18632/aging.202736

75. Nham GTH, Zhang X, Asou Y, Shinomura T. Expression of type II collagen and aggrecan genes is regulated through distinct epigenetic modifications of their multiple enhancer elements. Gene. 2019;704:134–141. doi:10.1016/j.gene.2019.04.034

76. Moon MH, Jeong JK, Lee YJ, Seol JW, Jackson CJ, Park SY. SIRT1, a class III histone deacetylase, regulates TNF-alpha-induced inflammation in human chondrocytes. Osteoarthritis Cartilage. 2013;21(3):470–480. doi:10.1016/j.joca.2012.11.017

77. Matsushita T, Sasaki H, Takayama K, et al. The overexpression of SIRT1 inhibited osteoarthritic gene expression changes induced by interleukin-1beta in human chondrocytes. J Orthop Res. 2013;31(4):531–537. doi:10.1002/jor.22268

78. Li Y, Xiao W, Wu P, et al. The expression of SIRT1 in articular cartilage of patients with knee osteoarthritis and its correlation with disease severity. J Orthop Surg Res. 2016;11(1):144. doi:10.1186/s13018-016-0477-8

79. Matsuzaki T, Matsushita T, Takayama K, et al. Disruption of Sirt1 in chondrocytes causes accelerated progression of osteoarthritis under mechanical stress and during ageing in mice. Ann Rheum Dis. 2014;73(7):1397–1404. doi:10.1136/annrheumdis-2012-202620

80. Qu ZA, Ma XJ, Huang SB, et al. SIRT2 inhibits oxidative stress and inflammatory response in diabetic osteoarthritis. Eur Rev Med Pharmacol Sci. 2020;24(6):2855–2864. doi:10.26355/eurrev_202003_20649

81. Zhu S, Donovan EL, Makosa D, et al. Sirt3 promotes chondrogenesis, chondrocyte mitochondrial respiration and the development of high-fat diet-induced osteoarthritis in mice. J Bone Miner Res. 2022;37(12):2531–2547. doi:10.1002/jbmr.4721

82. He Y, Wu Z, Xu L, et al. The role of SIRT3-mediated mitochondrial homeostasis in osteoarthritis. Cell Mol Life Sci. 2020;77(19):3729–3743. doi:10.1007/s00018-020-03497-9

83. Wang C, Yang Y, Zhang Y, Liu J, Yao Z, Zhang C. Protective effects of metformin against osteoarthritis through upregulation of SIRT3-mediated PINK1/Parkin-dependent mitophagy in primary chondrocytes. Biosci Trends. 2019;12(6):605–612. doi:10.5582/bst.2018.01263

84. Wang J, Wang K, Huang C, et al. SIRT3 activation by dihydromyricetin suppresses chondrocytes degeneration via maintaining mitochondrial homeostasis. Int J Biol Sci. 2018;14(13):1873–1882. doi:10.7150/ijbs.27746

85. Xu K, He Y, Moqbel SAA, Zhou X, Wu L, Bao J. SIRT3 ameliorates osteoarthritis via regulating chondrocyte autophagy and apoptosis through the PI3K/Akt/mTOR pathway. Int J Biol Macromol. 2021;175:351–360. doi:10.1016/j.ijbiomac.2021.02.029

86. Dai Y, Liu S, Li J, et al. SIRT4 suppresses the inflammatory response and oxidative stress in osteoarthritis. Am J Transl Res. 2020;12(5):1965–1975.

87. Liu H, Rosol TJ, Sathiaseelan R, Mann SN, Stout MB, Zhu S. Cellular carbon stress is a mediator of obesity-associated osteoarthritis development. Osteoarthritis Cartilage. 2021;29(9):1346–1350. doi:10.1016/j.joca.2021.04.016

88. Ji ML, Jiang H, Li Z, et al. Sirt6 attenuates chondrocyte senescence and osteoarthritis progression. Nat Commun. 2022;13(1):7658. doi:10.1038/s41467-022-35424-w

89. Chen J, Chen S, Cai D, Wang Q, Qin J. The role of Sirt6 in osteoarthritis and its effect on macrophage polarization. Bioengineered. 2022;13(4):9677–9689. doi:10.1080/21655979.2022.2059610

90. Ailixiding M, Aibibula Z, Iwata M, et al. Pivotal role of Sirt6 in the crosstalk among ageing, metabolic syndrome and osteoarthritis. Biochem Biophys Res Commun. 2015;466(3):319–326. doi:10.1016/j.bbrc.2015.09.019

91. Korogi W, Yoshizawa T, Karim MF, et al. SIRT7 is an important regulator of cartilage homeostasis and osteoarthritis development. Biochem Biophys Res Commun. 2018;496(3):891–897. doi:10.1016/j.bbrc.2018.01.129

92. Jensen ED, Schroeder TM, Bailey J, Gopalakrishnan R, Westendorf JJ. Histone deacetylase 7 associates with Runx2 and represses its activity during osteoblast maturation in a deacetylation-independent manner. J Bone Miner Res. 2008;23(3):361–372. doi:10.1359/jbmr.071104

93. Rossetto D, Avvakumov N, Cote J. Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics. 2012;7(10):1098–1108. doi:10.4161/epi.21975

94. Maeda K, Yoneda M, Nakagawa T, et al. Defects in centromeric/pericentromeric histone H2A T120 phosphorylation by hBUB1 cause chromosome missegregation producing multinucleated cells. Genes Cells. 2018;23(10):828–838. doi:10.1111/gtc.12630

95. Cao X, Wang X, Zhang W, et al. WNT10A induces apoptosis of senescent synovial resident stem cells through Wnt/calcium pathway-mediated HDAC5 phosphorylation in OA joints. Bone. 2021;150:116006. doi:10.1016/j.bone.2021.116006

96. Zhou X, Jiang P, Tan H, Wang Y, Bai L. KLF9-GRK5-HDAC6 axis aggravates osteoarthritis pathogenesis by promoting chondrocyte extracellular matrix degradation and apoptosis. Commun Biol. 2025;8(1):23. doi:10.1038/s42003-025-07460-x

97. Xie Y, Hu H, Liu M, et al. The role and mechanism of histone lactylation in health and diseases. Front Genet. 2022;13:949252. doi:10.3389/fgene.2022.949252

98. Zhang X, Wu Y, Pan Z, et al. The effects of lactate and acid on articular chondrocytes function: implications for polymeric cartilage scaffold design. Acta Biomater. 2016;42:329–340. doi:10.1016/j.actbio.2016.06.029

99. Wu J, Hu M, Jiang H, et al. Endothelial cell-derived lactate triggers bone mesenchymal stem cell histone lactylation to attenuate osteoporosis. Adv Sci. 2023;10(31):e2301300. doi:10.1002/advs.202301300

100. Maly K, Andres Sastre E, Farrell E, Meurer A, Zaucke F. COMP and TSP-4: functional roles in articular cartilage and relevance in osteoarthritis. Int J Mol Sci. 2021;22(5):2242. doi:10.3390/ijms22052242

101. Hu X, Huang J, Li Z, et al. Lactate promotes microglial scar formation and facilitates locomotor function recovery by enhancing histone H4 lysine 12 lactylation after spinal cord injury. J Neuroinflammation. 2024;21(1):193. doi:10.1186/s12974-024-03186-5

102. Han S. Osteoarthritis year in review 2022: biology. Osteoarthritis Cartilage. 2022;30(12):1575–1582. doi:10.1016/j.joca.2022.09.003

103. Huh YH, Ryu JH, Chun JS. Regulation of type II collagen expression by histone deacetylase in articular chondrocytes. J Biol Chem. 2007;282(23):17123–17131. doi:10.1074/jbc.M700599200

104. Qu H, Li J, Wu LD, Chen WP. Trichostatin A increases the TIMP-1/MMP ratio to protect against osteoarthritis in an animal model of the disease. Mol Med Rep. 2016;14(3):2423–2430. doi:10.3892/mmr.2016.5523

105. Chen WP, Bao JP, Hu PF, Feng J, Wu LD. Alleviation of osteoarthritis by Trichostatin A, a histone deacetylase inhibitor, in experimental osteoarthritis. Mol Biol Rep. 2010;37(8):3967–3972. doi:10.1007/s11033-010-0055-9

106. Zhang H, Ji L, Yang Y, et al. The therapeutic effects of treadmill exercise on osteoarthritis in rats by inhibiting the HDAC3/NF-KappaB pathway in vivo and in vitro. Front Physiol. 2019;10:1060. doi:10.3389/fphys.2019.01060

107. Cheng C, Shan W, Huang W, et al. ACY-1215 exhibits anti-inflammatory and chondroprotective effects in human osteoarthritis chondrocytes via inhibition of STAT3 and NF-kappaB signaling pathways. Biomed Pharmacother. 2019;109:2464–2471. doi:10.1016/j.biopha.2018.11.017

108. Feige JN, Lagouge M, Canto C, et al. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 2008;8(5):347–358. doi:10.1016/j.cmet.2008.08.017

109. Nishida K, Matsushita T, Takayama K, et al. Intraperitoneal injection of the SIRT1 activator SRT1720 attenuates the progression of experimental osteoarthritis in mice. Bone Joint Res. 2018;7(3):252–262. doi:10.1302/2046-3758.73.BJR-2017-0227.R1

110. Leah E. Osteoarthritis: chondroprotection by histone deacetylase inhibition. Nat Rev Rheumatol. 2013;9(6):321. doi:10.1038/nrrheum.2013.63

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.