
Epigenetic roles of chromatin remodeling complexes in bone biology and the pathogenesis of bone‑related disease (Review)
- Authors:
- Published online on: May 28, 2025 https://doi.org/10.3892/ijmm.2025.5556
- Article Number: 115
-
Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Bone is a highly dynamic tissue that undergoes continuous remodeling to maintain skeletal integrity and support various physiological functions (1,2). This remodeling process, known as bone turnover, involves a delicate balance between bone formation and resorption (3). Through this ongoing process, bone homeostasis is achieved, which is critical for maintaining overall skeletal health. Specifically, bone homeostasis is maintained through the coordinated activities of osteoblasts, osteoclasts, osteocytes and chondrocytes, which work together to ensure that bone density and structure remain stable (4). Disruptions in this balance can lead to various bone-related diseases. Bone-related diseases encompass a variety of conditions such as osteoporosis, osteopetrosis, osteonecrosis, osteoarthritis (OA), osteosarcoma and chondrosarcoma, characterized by abnormalities in bone density, structure and integrity (5). These conditions can lead to grave corresponding clinical symptoms. For instance, osteoporosis is characterized by increased bone loss and low peak bone mass, which give rise to low bone density (bone brittleness); with a propensity to fall and poor bone quality, fractures are inexorable in this disease (6). Additionally, osteopetrosis manifests with higher bone mass resulting in phenotypic features such as macrocephaly and altered craniofacial morphology, especially influencing other organs and tissues, notably the bone marrow and nervous system (7). While OA is a complex condition involving mechanical cartilage degradation and matrix protease activities, resulting in pain and loss of joint function, osteonecrosis occurs when there is a lasting inflammatory environment due to distinguished detrimental factors such as glucocorticoids or alcohol, triggering bone tissue death (8,9). As for bone tumors, they are rare and heterogeneous groups of neoplasms that occur in the bones (10). Epigenetic research has greatly enhanced our understanding of these diseases by focusing on gene expression regulation rather than changes in the DNA sequence itself (11). Key epigenetic mechanisms involve DNA methylation, histone modifications, non-coding RNAs (ncRNAs) and chromatin remodeling complexes (12). For instance, ncRNAs, including microRNAs (miRNAs) and long ncRNAs (lncRNAs), play crucial roles in regulating bone formation, resorption processes and tumorigenesis in bone cancer (13). These epigenetic factors collectively impact the differentiation, proliferation and apoptosis of bone cells, contributing to the development and progression of bone-related diseases (14-19).
Chromatin is a densely packed and highly regulated structure that packages DNA, facilitating numerous cellular processes in eukaryotes, particularly in the context of gene expression regulation, DNA replication and repair. The fundamental unit of chromatin is the nucleosome, a highly stable structure that wraps 145-147 bp of DNA around an octamer of histone proteins (20). The 2-meter-long DNA is compressed into chromatin within the nucleus of each cell, creating a notable barrier to cellular processes such as transcription (21). Chromatin remodeling complexes are sophisticated multiprotein assemblies that serve as pivotal regulators of chromatin architecture, mediating dynamic changes that profoundly influence gene expression, DNA repair, replication and a wide range of other critical cellular processes (22). To date, four families of chromatin remodeling complexes have been identified: Switch/sucrose-non-fermenting (SWI/SNF), imitation switch (ISWI), chromodomain-helicase-DNA binding (CHD) and inositol requiring 80/SWi2/snf2-related 1 (INO80/SWR) (23). Each of these families perform unique, non-redundant roles within the cell. Chromatin remodeling complexes are typically composed of three parts: Nucleosome, module [ATPase module, actin-related protein (ARP) module or body module] and proteins. However, different chromatin remodeling complexes have notably different components, including diversities in the nucleosomes and configurations. For example, the SWI/SWF family is composed of the nucleosome, ATPase module, ARP module, body module and diverse proteins (including snf2, RT102, Arp7 and Arp9) (24). The ISWI family does not entail an ARP module and contains dissimilar proteins, such as Isw1, Ioc2, Ioc3 and Ioc4 (25). For the CHD family, the ATPase module exclusively contains distinct proteins. SWR1 contains three main modules and differing proteins (26). Therefore, with such a framework, chromatin remodeling complexes comprise crucial enzymes or cooperate with other chromatin-related factors to play a key role in dynamically regulating various cellular processes, including transcription and DNA repair, by controlling access to genomic DNA (26). Additionally, these gene-mediated chromatin remodeling complexes are crucial contributors to the molecular pathology of bone-related diseases by engaging in bone epigenetic mechanisms such as differentiation, proliferation and senescence. For instance, disturbances in bromodomain-containing protein (BRD)9 within the SWI/SWF family can lead to debilitating bone-related diseases such as osteoporosis, osteopetrosis and osteonecrosis (14).
Therefore, chromatin remodeling complexes play a pivotal role in bone-related diseases. The present review aims to provide an overview of the mechanisms and functions of gene-mediated chromatin remodeling complexes in bone-related diseases. Through literature mining and investigation (Data S1), the present review will examine the relationship between chromatin remodeling complexes and bone-related diseases, and the potential clinical relevance of this relationship, such as pathological mechanisms and disease targets, ultimately aiming to generate further effective clinical therapeutic applications, such as BRM inhibitors for osteoporosis treatment and BRD9 inhibitors for advanced synovial sarcoma treatment. The primary strength of the present review lies in its thorough examination of the role of chromatin remodeling complexes specifically in bone-related diseases. Additionally, the present review emphasizes the potential therapeutic applications of chromatin remodeling complexes, which has been a relatively underexplored area in bone research. Unlike earlier reviews (27-30), which tend to focus on a narrow subset of chromatin remodeling complexes or bone diseases, the present review covers a broader range of chromatin remodeling complexes and bone-related diseases, including bone tumors, OA, bone-fat disorder, adolescent idiopathic scoliosis (AIS) and osteoporosis. A detailed review of the role and mechanisms of chromatin remodeling complexes in bone metabolic processes, such as stemness maintenance, chondrogenesis, osteogenesis, osteoclastogenesis and angiogenesis, are also provided. Additionally, the impact of chromatin remodeling complexes on key bone biological functions, including proliferation, senescence and programmed cell death are explored in the present review. Another strength of this review is its focus on therapeutic implications, such as how chromatin remodeling complexes could be targeted for intervention in bone-related diseases.
Classification, mechanisms and regulations of chromatin remodeling complexes
Chromatin remodeling complexes primarily operate through three fundamental mechanisms (Fig. 1). First, these complexes can edit assembled nucleosomes by replacing, moving or removing specific nucleosomes, thus altering chromatin structure and function. Second, chromatin remodeling complexes are involved in the assembly and reorganization of nucleosomes from random deposition to regular space. Third, chromatin remodeling complexes alter the structure of chromatin to make specific regions of DNA more accessible to transcription factors and other regulatory proteins (22,31,32). This is achieved through various mechanisms, including repositioning nucleosomes, nucleosome ejection and dimer ejection, which all contribute to the dynamic remodeling of chromatin (22,31,32). Collectively, these mechanisms serve as the driving force behind chromatin activation and represent a key component of the hereditary code governing diverse epigenetic regulations.
Based on the sequence homology of the catalytic ATPase and the accessory subunits, chromatin remodeling complexes are classified into four distinct families (Fig. 2): SWI/SNF, ISWI, CHD and INO80/SWR (33,34). The SWI/SNF complex in human cells is characterized by a notable diversity of assemblies. There are several variants of this complex, including canonical BAF (cBAF), polybromo BAF (PBAF) and non-canonical BAF (ncBAF/gBAF), each distinguished by the presence of unique subunits (26). However, the ISWI and CHD family have only one assembly (22). Although INO80 and SWR belong to the same family of chromatin remodelers, they are classified into different subfamilies with distinct components (22,35,36). To this point, each family of chromatin remodeling complexes performs specialized functions within the cell (26). The SWI/SNF family, for instance, includes the BRG1 ATPase subunit, which can independently remodel nucleosome positioning on nucleosomal templates. A minimal four-subunit complex composed of BRG1, BAF155, BAF170 and BAF47 efficiently achieves chromatin remodeling in vitro (37). ISWI complexes facilitate the maturation of initial histone-DNA complexes (pre-nucleosomes) into canonical octameric nucleosomes and help space nucleosomes at relatively fixed distances (38,39). CHD family members contain a conserved ATPase domain, comprising SNF2_N and Helicase_C PFAM domains, which act as a motor to drive dynamic interactions with chromatin and nucleosome substrates (40,41). However, INO80, features multiple ATPases that provide catalytic activity and enable ATP-dependent nucleosome sliding within the cell (42,43). Additionally, Ino80p serves as a docking site for several core subunits, including actin, ARPs and the Rvb AAA+ ATPases (44). While these chromatin remodeling complex families implement diverse functions in vivo, they share a common mission: Facilitating genome utilization, which includes processes such as transcription factor binding, DNA replication and repair (45,46). It is important to note that ATP-dependent chromatin remodeling complexes can be distinguished not only by the composition of their subunits but also by the domain organization of their catalytic subunits (32). Overall, chromatin remodeling complexes play a crucial role in epigenetic regulation.
Chromatin remodeling complexes are regulated by several factors that govern their own ultimate epigenetic functions (Fig. 3). First, ncRNAs, miRNAs and lncRNAs exert a pivotal influence on chromatin remodeling complexes. miRNAs can target the mRNA of chromatin remodeling complex subunits pre-transcriptionally, leading to their degradation, and can also inhibit the translation of these subunits. lncRNAs can interact with chromatin remodeling subunits post-transcriptionally in several ways including binding to these subunits to inhibit the assembly of the complex, acting as guiding lncRNAs to recruit chromatin remodeling complexes and as decoy lncRNAs, limiting the chromatin localization of the complexes (47-51). Second, actin and ARPs are integral components of chromatin remodeling complexes and regulate ATPase activity, facilitate complex assembly and enhance DNA binding and nucleosome repositioning through direct interaction with ATPase subunits. Additionally, ARPs interact with histones and serve as molecular bridges between chromatin-modifying complexes, coordinating higher-order chromatin regulation (22,52). Lastly, chromatin remodeling complexes are under the strict regulation of posttranslational modifications, namely, they are modulated by phosphorylation, acetylation and PARylation (22,53,54). For example, during mitosis, human SWI/SNF is phosphorylated and this modification inhibits its remodeling activity, which may contribute to the widespread inhibition of chromatin remodeling during cell division (54). What's more, the human nucleosome acetyltransferase of the histone H4 (NuA4)/Tat-interactive protein 60 kDa coactivator complex, a homologous fusion of the yeast SWR1 and NuA4 complexes, facilitates the incorporation of the histone variant H2A.Z into nucleosomes while acetylating histones H4, H2A and H2A.Z. This dual function plays a crucial role in gene regulation and genome stability maintenance (52). Additionally, Sala et al (55) found that PARylated ISWI displays a reduced nucleosome-binding affinity, as well as lower DNA- and nucleosome-stimulated ATPase activity. Therefore, chromatin remodeling complexes must first be regulated by upstream modulators before exerting their functions. Under normal conditions, chromatin remodeling complexes serve as facilitators and protectors of cellular molecular activities, but aberrations in their functions can lead to various diseases, such as cancer, Coffin-Siris syndrome and fibromuscular dysplasia (56). Chromatin remodeling complexes play a pivotal role in bone pathology (11,28). Numerous studies have identified a variety of relevant chromatin remodeling complexes and their subunits as well as elucidated their specific functions (26,37). For instance, the SWI/SWF complex includes subunits such as BRG1, BRM, BAF45, BAF47, BAF155, BAF170, BAF180, BAF200, BAF250a, BRD7, BRD9 and SS18. The ISWI complex is characterized by the presence of the bromodomain adjacent to zinc finger domain 1A (BAZ1A) subunit. The CHD family comprises subunits such as CHD1, CHD4, CHD7 and CHD9, and the INO80/SWR complex is composed of multiple subunits, such as INO80, SWR1, ARP4, ARP5, RvB1 and RvB2 that participate in chromatin remodeling. Each subunit is involved in distinct functions, with some overlapping roles and others being unique (28). The relationship between chromatin remodeling complexes and bone biology and bone-related diseases is dissected further below.
Role of chromatin remodeling complexes in bone metabolic processes
Chromatin remodeling complexes are intimately linked to bone metabolic processes, performing various regulatory mechanisms and functions in the modulation of gene expression (Figs. 4 and 5). For instance, their expression at specific stages of bone biology directs progenitor cells to differentiate and proliferate into osteoblasts. Conversely, these complexes can also contribute to bone-related diseases when their functions become aberrant (28,29,57). Therefore, in this section, the specific functional properties of these chromatin remodeling complexes in normal bone metabolic processes as well as aberrant settings are discussed (Table I).
![]() | Table IDiverse role of chromatin remodeling complexes in the bone metabolic process and the biological functions of bone. |
Chromatin remodeling complexes and stemness maintenance
Stemness maintenance is crucial for bone development and the prevention of bone-related diseases, as it ensures the continuous supply of progenitor cells capable of differentiating into osteoblasts and other bone-forming cells (58,59). In the following sections, the specific mechanisms through which chromatin remodeling complexes influence stemness maintenance, and their implications for bone biology and disease, will be discussed.
BRG1 (also known as SMARCA4) and BRM (also known as SMARCA2)
Well-established interactions between esBAF (BRG1 or BRM) and polycomb complexes have been documented, highlighting that both esBAF and polycomb group proteins play roles in maintaining pluripotency through both antagonistic and synergistic acts in polycomb group proteins (60,61). In osteoblast precursors, BRG1-containing SWI/SNF typically induces the expression of osteogenic genes, while BRM-specific complexes act in conjunction with p130 and repressor E2Fs to primarily restrain differentiation. This action helps sustain the committed precursor state until pre-osteoblasts receive the appropriate differentiation signals (60). Moreover, BRG1 and BRM can function as either oncogenes or tumor suppressors. In chondrosarcoma, both subunits in the esBAF complex exhibit oncogenic properties when inhibited (62). Conversely, loss of BRG1 leads to a loss of both self-renewal capacity and pluripotency (62,63), while the loss of BRM results in enhanced osteoblast differentiation and impaired adipocyte differentiation (64). Additionally, a previous study found that BRG1 interacts with Ewing sarcoma-friend leukemia integration 1 (EWS-FLI1) fusion protein or Ewing sarcoma RNA binding protein 1 (EWSR1), with the EWS-FLI1 fusion protein being detected in >85% of Ewing sarcoma cases. This interaction is a key regulator in the cell growth of Ewing sarcoma (65).
BAF47 (also known as SMARCB1/INI1)
BAF47 plays a crucial role in ensuring the stability and functionality of the BAF complex. Specifically, the presence of BAF47 is essential for the SWI/SNF complex to effectively resolve bivalency in the genome, highlighting its critical role in regulating gene expression at bivalent promoters and enhancers (66,67). Therefore, the loss of BAF47 decreases the chromatin affinity of the intact BAF complex, leading to dysfunctional BAF complex activity, which is vital for every step of bone development (67). A study demonstrated that knockdown of BAF47 impedes differentiation in human embryonic stem cells (hESCs), while upregulation of BAF47 enhances differentiation (68,69). Therefore, BAF47 plays a notable role in maintaining the balance of stemness in the human body.
BAF155 (also known as SMARCC1) and BAF170 (also known as SMARCC2)
Together with BRG1, BAF155 and BAF170 play pivotal roles in maintaining pluripotency and self-renewal in hESCs. The depletion of these three subunits leads to pluripotency dysfunction in hESCs, significantly reducing the potential for future bone development (62,70). Additionally, BAF155 interacts with EWS-FLI1, which acts as an aberrant transcription factor that drives the cellular transformation of Ewing sarcoma. Moreover, this chimeric protein directly binds to GGAA repeats, thereby modifying the transcriptional profile of Ewing sarcoma, eventually promoting the progression of this disease (65).
BRD9
BRD9 is recruited by BRD4 to regions linked to naive pluripotency genes in a bromodomain-dependent manner, playing a crucial role in regulating and maintaining the naive pluripotent state of ESCs (71). Inhibition of BRD9 expression leads to decreased levels of NANOG and SOX2, impairing self-renewal and altering differentiation, particularly inhibiting mesoderm differentiation while promoting neural ectoderm differentiation. Since the mesoderm is essential for the development of various tissues, including cartilage and bone, its disruption can affect skeletal formation (72). BRD9 also regulates pluripotency gene expression and TGF-β/Nodal/Activin and Wnt signaling pathways by forming a complex with BRD4, SMAD2/3, β-CATENIN and p300, and modulating H3K27 acetylation to sustain hESC differentiation (72). Furthermore, inhibition of BRD9 enhances somatic cell reprogramming by downregulating fibroblast genes, limiting chromatin accessibility at enhancers and reducing reprogramming inhibitors such as MN1 and ZBTB38, thereby facilitating reprogramming (73).
CHD1
CHD1 is an ATPase-dependent chromatin remodeling protein that plays a crucial role in maintaining pluripotency and stemness in ESCs and mesenchymal stem cells (MSCs) by facilitating chromatin accessibility and regulating key gene expression. hESCs possess chromatin accessibility that enables them to differentiate into various daughter cell types, including hematopoietic, neural stem, mesenchymal stem and other lineage cells (74-76). Notably, MSCs serve as progenitor cells of osteoblasts, and osteoprogenitors originating from mesenchymal stromal cells are also committed to their differentiation into osteoblasts, the cells responsible for bone formation and maintenance (77,78). In this context, CHD1 plays a critical role in the mediator complex, facilitating chromatin accessibility in hESCs, pre-initiating gene transcription and inducing pluripotent stem cells by maintaining a heterochromatin-poor pluripotent stem cell state (75). Specifically, CHD1 is crucial for maintaining open chromatin and pluripotency in mouse ESCs; its downregulation leads to heterochromatin accumulation, loss of pluripotency and the impaired reprogramming of somatic cells to the pluripotent state (74). Moreover, CHD1 is essential for early mouse embryogenesis and pluripotency, as its loss results in embryonic lethality after implantation by significantly downregulating key regulators of cell fate specification, such as Pou5f1 and Nanog, and impairing the activation of Hmgpi during zygotic gene activation, which can be rescued by Hmgpi mRNA microinjection (79). Furthermore, CHD1 is vital for maintaining the integrity of the genome in ESCs by preventing the accumulation of DNA double-stranded breaks, thereby further supporting their pluripotency and overall stemness during hypertranscription (76). Additionally, a previous study demonstrated that CHD1 regulates the stemness of MSCs, with higher levels promoting their tissue regeneration and hematopoietic stem cell-supporting activity, while lower levels are associated with a loss of clonogenic potential in pathological conditions (2). Ultimately, the loss of CHD1 leads to the transformation of heterochromatin and a significant loss of pluripotency (28,80).
CHD7
CHD7 is a crucial gene for stemness maintenance as it regulates epigenetic and signaling pathways and is highly expressed in active gene expression signals within stem cells (80). Research has shown that CHD7 is also vital for maintaining open chromatin in ESCs, regulated by the trithorax protein Ash2l, and the knockdown of Ash2l reduces H3K4 methylation and promotes a silenced chromatin state (81) Additionally, the co-localization of BRG1 and CHD7 at distal regions in ESCs influences the expression of key transcription factors such as NANOG, SOX2 and OCT4, further affecting ESC pluripotency (82). Furthermore, it has been reported that CHD7 interacts with lysine-specific histone demethylase 1 (LSD1) in mouse ESCs and is crucial for maintaining stemness and proper differentiation. The increased co-occupancy of methylated H3K4 and CHD7 on chromatin following LSD1 deletion underscores the essential role of LSD1 in facilitating the binding of CHD7 to chromatin and regulating differentiation (83). Finally, CHD7 knockout ESCs exhibit defective expression of ectodermal markers that affect pluripotency, which is vital for the stemness associated with subsequent differentiation into bone, cartilage and skeletal muscle (83). Notably, the depletion of CHD7 in MSCs leads to the repression of osteogenic transcription factors and impairs the osteogenesis ability of these cells (84).
CHD9
CHD9 is dispensable for pluripotent marker expression, including OCT4, SOX2, NANOG and SSEA-1 (85). The knockout of CHD9 promotes hESC proliferation through binding to genomic domains that overlap with DNA motifs recognized by cell cycle-related transcription factors and interacting with the transcriptional regulation of Pol I-controlled genes and nuclear receptors (85,86). Additionally, a study found that CHD9 is transiently expressed during mesenchymal cell differentiation both in vivo and in vitro, with expression observed in osteoprogenitors and downregulation in mature osteoblasts (78).
INO80
The INO80 is crucial for the self-renewal of hESCs, as well as for reprogramming and embryonic development. INO80 specifically binds to the promoters of key pluripotency genes in conjunction with transcription factors such as OCT4, NANOG, SOX2 and Krüppel-like factor 4, with its binding reliant on WD repeat domain 5 (WDR5). INO80 helps maintain an open chromatin structure, enabling the recruitment of the mediator complex and RNA polymerase II, which enhances the expression of these pluripotency genes (87). Furthermore, silencing INO80 leads to the abnormal morphology of hESCs (87). While in mouse ESCs, INO80 deficiency causes a notable increase in the duration of the G1-phase in ESCs cultured under primed conditions. Additionally, INO80 directly associates with the transcription start site to modulate the expression of genes involved in the cell cycle, including Ccne1, Cdc25b and Cdkn1b (78).
Chromatin remodeling complexes and chondrogenesis
Chondrogenesis is the process by which MSCs differentiate into chondrocytes, ultimately forming cartilage models (anlagen) that serve as precursors to future bones through endochondral ossification while also secreting cartilage matrix to form cartilage (88,89). In the following section, the impact of chromatin remodeling complexes on chondrogenesis under both normal and pathological conditions of cartilage will be discussed.
BRG1
BRG1 is an indispensable chromatin regulator that engages in numerous cellular activities and plays a core role in DNA replication, repair, recombination and transcriptional regulation (90,91). The expression of fibroblast growth factor receptor 3 (FGFR3) is essential for cartilage development and can be induced by bone morphogenic protein (BMP) 2. This induction is mediated by the downstream factor Sp1, which is influenced by BRG1. BRG1 promotes FGFR3 expression by remodeling chromatin regions that have Sp1-binding sites at the FGFR3 transcriptional initiation sites. This alteration enhances Sp1's binding to the proximal promoter, which facilitates p300 recruitment and leads to modifications in the 'histone code', including phosphorylation and methylation changes (92). The natural small molecule spermidine has been reported to protect against and restore cartilage damage in OA (93-95), and BRG1 mediates its protective effects by enhancing osteoarthritic cartilage through the Nrf2/KEAP1 and STAT3 signaling pathways (96).
BAF47
Studies have shown that BAF47 serves as a genetic hallmark in malignant tumors, and its loss is associated with extraskeletal myxoid chondrosarcoma (EMC) featuring rhabdoid cytological characteristics. However, in one case of EMC exhibiting high-grade features such as increased cellularity and prominent nucleoli, with no rhabdoid features present, the expression of nuclear BAF47 remained intact (97,98). Additionally, BAF47 levels are reduced in synovial sarcoma, and this reduction can be assessed using immunohistochemical staining for diagnostic purposes (29,99). In addition, compared with other atypical teratoid tumors where BAF47 is absent or lost, in mesenchymal chondrosarcoma, the level of BAF47 is retained (100). Therefore, BAF47 is an excellent marker to diagnose different tumor types but the mechanisms remain unclarified (101).
SS18
Mutations in the BAF subunit across human cancer types display a notable tissue-specific pattern, with BAF subunits mutated in >20% of all human cancers (102). Notably, almost all cases of synovial sarcoma are attributed to the SS18-SSX fusion due to the t(X;18) translocation, whereas mutations in the SS18 BAF subunit are infrequently observed in other types of cancer (103,104). The underlying mechanism involves the SS18-SSX gene fusion, which indirectly inactivates the function of SMARCB1 by binding to the chromatin remodeling complex, thereby promoting the transcription of the oncoprotein, SS10-SSX, and contributing to the development of synovial sarcoma (104,105).
BAF155
BAF155 is a component of SWI/SNF chromatin remodeling complexes, which help make DNA more accessible by altering the arrangement of nucleosomes during gene transcription (106). The targeted deletion of BAF155 leads to a failure in the transcriptional activation of Sonic hedgehog (Shh)/Gli target genes within limb buds that are undergoing development, such as the Ptch1 receptor for Shh and its downstream effector Gli1 in the posterior region of the limb bud. This disruption of the Hh pathway is associated with aberrant BMP activity and the initiation of chondrogenesis (107).
Chromatin remodeling complexes and osteogenesis
Osteogenesis is the biological process through which new bone is formed, involving the differentiation of MSCs into osteoblasts (108). This complex process is regulated by various signaling pathways and factors, including hormones, growth factors, transcription factors and ncRNAs, ensuring proper bone development and homeostasis (109,110). Regulating MSC osteogenic differentiation is essential for advancing bone tissue engineering (111). Several subunits of chromatin remodeling complexes play a pivotal role in osteogenesis, making them valuable for applications in bone tissue engineering (84,112). For example, overexpression of CHD7 can interact with downstream factors of BMP signaling, such as SMAD1, thereby enhancing the osteogenic potential of MSCs (84). Additionally, BAF180 influences MSC osteogenic differentiation by modulating BMP/Smad signaling, improving chromatin accessibility, and increasing binding at key osteogenic genes, including Alpl, BmprIb, TgfbrII and Runx2, in BMP2-treated MSCs (112). Overall, a deeper understanding of how chromatin remodeling complexes regulate MSC osteogenic differentiation can optimize the ability of MSCs to differentiate into osteoblasts, thereby facilitating their application in bone tissue engineering and the treatment of bone-related diseases (84).
BRG1
BRG1, a core component of the SWI/SNF chromatin remodeling complex that functions as an ATPase, has been identified in developing skeletal structures of the mouse embryo and in ex vivo osteoblast cultures (113). A study has demonstrated that BRG1 expression is consistently present throughout skeletal components and is positively correlated with the key osteogenic regulatory protein, runt related transcription factor 2 (RUNX2) (57). These observations suggest that SWI/SNF chromatin remodeling activity is essential for supporting RUNX2-dependent skeletal gene expression. RUNX2 is crucial for initiating osteogenesis upstream of SP7 in the regulatory hierarchy of osteoblast development, ensuring independent and autonomous regulation in cartilage and bone development (57,114). In the context of BRG1 depletion, the induction of alkaline phosphatase, an osteogenic marker, is impaired in pre-osteoblasts (115). Additionally, another study found that the knockout of BRG1 severely damages the ability of bone cells to mineralize and to express key markers of osteoblast differentiation, such as osteocalcin and alkaline phosphatase (60). Osteocalcin itself plays an important role in determining bone size, shape and strength (116,117). Furthermore, BRG1-containing SWI/SNF complexes stimulate basal tissue and vitamin D3-enhanced osteocalcin promoter activity via the transcription factor, CCAAT/enhancer-binding protein β. This factor, together with RUNX2, forms a stable complex that facilitates RNA polymerase II binding and the activation of osteocalcin gene transcription in osteoblastic cells (118). Moreover, p107, alongside retinoblastoma protein (pRB), plays a distinct role in the induction of the osteogenic gene, Alpl, during osteoblast differentiation, with p107 being crucial for the efficient recruitment of the BRG1-SWI/SNF chromatin-remodeling complex, necessary for Alpl activation (119).
BRM
Notably, unlike other subunits of the chromatin remodeling complex, BRM-deficient preosteoblasts exhibit premature expression of osteoblast differentiation markers suggesting an enhancement of osteogenesis and subsequent bone formation (64,120).
BAF45a and BAF45d
In the BAF45 family, there are four homologs, including BAF45a, BAF45b, BAF45c and BAF45d (68,121). Among these homologs, BAF45a and BAF45d are more accessible during the induction of bone MSC differentiation into osteoblasts compared with BAF45b and BAF45c, as BAF45a and BAF45d are preferentially expressed in osteoblasts. Notably, BAF45a serves as a vital subunit of the chromatin regulatory complex and, through the bone tissue-specific RUNX2-BAF45A-EZH2 molecular epigenetic axis, determines chromatin accessibility during the early stage of commitment and differentiation of mineralized cells (122). The deletion of BAF45a results in decreased promoter accessibility for vital transcription factors required for the induction and maturation of osteoblasts (15,122).
BAF180 (also known as PBRM1)
Mechanistically, BMP and osteogenic signaling pathways induce the expression of BAF180 and PBAF in MSCs, promoting ossification in vivo by affecting the activation of SMAD1/5/8 through locus-specific epigenomic remodeling. This involves PBRM1 bromodomains and the transcriptional regulation of BMPR/TGFβRII, which together lead to the expression of BMP-early-responsive genes and facilitate the progression of osteogenesis (57,112).
BAF200 (also known as ARID2)
A study has suggested that BAF200 is crucial for promoting osteoblast commitment and differentiation (123). BAF200 targets key osteogenic markers, such as the Alpl promoter and Bglap, and in some cases, significantly contributes to the recruitment of RNA polymerase II, ultimately facilitating the commitment and differentiation of osteoblasts. Conversely, the depletion of BAF200 expression during the osteoblast differentiation process results in a failure to mineralize into a calcified bone matrix (124,125).
BRD7
In addition to functioning alongside BAF180 and BAF200 as components of the PBAF complex to promote osteogenesis (112), BRD7 has also been identified as a novel substrate of the APC/C-E3 ligase during the cell cycle. This role may provide a therapeutic target for treating osteosarcoma and serves as a crucial regulator for stabilizing and suppressing tumor progression in osteosarcoma (126).
CHD1
As a member of the CHD family, CHD1 is required for the induction of osteoblast-specific gene expression, extracellular matrix (ECM) mineralization and ectopic bone formation in vivo. CHD1 is closely associated with transcription and nucleosome turnover downstream of the transcriptional start site (TSS); it exhibits high occupancy around the TSS of differentiation-activated genes, thereby supporting the differentiation of MSCs into osteoblasts (2,75).
CHD7
SMAD1 and SP7 are core components of the canonical BMP signaling pathway. Previous studies have elucidated that CHD7 is required for the osteogenic differentiation of human bone MSCs by interacting with SMAD1 and binding to the enhancer region of SP7 to promote differentiation. By contrast, the absence of CHD7 impairs bone formation in vivo (80,84). Additionally, other studies have demonstrated that depletion of CHD7 in MSCs and preosteoblasts results in a phenotype characterized by low bone mass and significantly increased marrow adiposity. This occurs through the enhancement of the peroxisome proliferator-activated receptor (PPAR) signaling associated with methylated H3K4 patterns, which subsequently activates the transcription of downstream lipogenic genes. This disruption of the balance between osteogenesis and lipogenesis ultimately results in bone and fat disorder (127-129).
CHD9
CHD9, a master regulator of ribosomal gene transcription in MSCs and osteogenesis, works synergistically with BRG1 to upregulate the expression of RUNX2 (86,130). Additionally, CHD9 binds to skeletal tissue-specific promoters associated with key genes involved in osteoblast differentiation, including glycan, core-binding factor subunit α1, collagen II, osteocalcin and myosin (85).
INO80
The only subunit in the INO80/SWR family that contributes to osteogenesis is INO80. INO80 interacts with WDR5 in MSCs, regulates canonical Wnt activity and stabilizes the expression of key osteogenic markers, including RUNX2, SP7, collagen type I α1 chain and osteopontin, all of which promote osteogenesis (131).
Chromatin remodeling complexes and osteoclastogenesis
Osteoclastogenesis plays a crucial role in bone development and bone diseases by regulating the resorption of bone tissue, thereby maintaining the balance between bone formation and degradation (132). On the other side of the bone remodeling balance, osteoclastogenesis requires chromatin remodeling complexes to exert their influence and contribute to the overall process.
BAF250a (also known as ARID1a/OSA1)
The expression of BAF250a facilitates the cooperation between chromatin and the transcription factor, Jun/Fos, leading to the upregulation of sialic acid-binding Ig-like lectin 15 in osteoclast precursors, thereby triggering the differentiation of osteoclasts (16). Moreover, BAF250a plays a critical role in safeguarding osteoclast fate canalization during the proliferation-differentiation switch by facilitating the formation of transcriptional condensates with coactivator BRD4 and lineage-specifying transcription factor PU.1 at the Nfatc1 super-enhancer (133). Additionally, the antagonistic interactions between the ARID1A-cBAF and BRD9-ncBAF complexes, along with the dependency on coactivator BRD4, highlight the intricate balance required for proper cell fate canalization during osteoclastogenesis (133). Furthermore, a recent study has shown that the expression of BAF250a is downregulated in osteosarcoma compared with non-tumor tissues, suggesting its potential as a novel prognostic and therapeutic marker (134,135).
BRD9
BRD9 is upregulated during receptor activator of NF-κB ligand-induced osteoclast differentiation and suppresses osteoclastogenesis by interacting with the transcription factor, FOXP1. This interaction enhances STAT1 transcription by increasing chromatin accessibility at the STAT1 promoter and enhancer regions, subsequently activating IFN-β signaling, which serves as a negative feedback mechanism for osteoclastogenesis. Conversely, the absence of BRD9 expression can lead to debilitating bone diseases such as osteoporosis, osteopetrosis, osteonecrosis and Paget's disease (14).
Chromatin remodeling complexes and angiogenesis
Angiogenesis is crucial for bone formation and resorption, as blood vessels play a vital role in bone regeneration and are influenced by aging and pathological conditions (136,137). Vascular endothelial growth factor (VEGF), which is expressed by osteoprogenitors, hypertrophic chondrocytes and osteoblast precursors, regulates angiogenesis within the skeletal system (138). In addition, the hypoxia-inducible factor (HIF)-1α pathway is essential for maintaining bone homeostasis and angiogenesis, significantly impacting the development of bone metabolic diseases (138). Sena et al (139) demonstrated that BRM and BRG1 enhance the hypoxic induction of HIF1α and HIF2α genes, along with their target genes, by recruiting BRG1 complexes to gene promoters. Additionally, the SWI/SNF subunits, BRM and BRG1, enhance HIF-1-mediated target gene activation in an ATPase-dependent manner and are recruited to the VEGF gene promoter under hypoxia (140). CHD4 also acts as a crucial coactivator of HIF1 and HIF2, amplifying HIF-driven transcriptional programs (141). Although there is currently no direct research exploring the role of chromatin remodeling complexes in angiogenesis during bone development and related diseases, their influence on angiogenesis-related genes suggests that they may also play a significant role in these processes.
During the embryonic stage, angiogenesis serves a crucial role in bone development by providing essential signaling molecules, such as VEGF, and supporting osteoblasts and bone marrow stromal cells. This process not only promotes bone formation but also contributes to bone function, making it a key regulatory factor in bone development (142). It has been reported that BRG1-deficient mutants die by embryonic day 11.5 and show abnormal angiogenesis in the yolk sac (143). BRG1 deletion also reduces COUP-TFII expression in venous endothelial cells and causes improper arterial marker expression in veins during embryonic vascular development (144). CHD4 also plays a key role in embryonic vasculature development by suppressing plasmin activation, with its loss leading to increased urokinase-type plasminogen activator receptor expression and decreased thrombospondin-1, both critical for plasmin activation (145). While chromatin remodeling complexes play a crucial role in embryonic vascular development, their influence on angiogenesis during bone formation and its overall impact on bone development remain largely unexplored. Further research is needed to deepen the understanding of these mechanisms and their relevance to bone-related diseases.
Role of chromatin remodeling complexes in bone biological functions
Normal bone development and aging are regulated by a complex interplay of biological and physiological processes that work in concert to sustain skeletal health throughout the lifespan (146). This section mainly focuses on chromatin remodeling complexes in three notable aspects: Proliferation, senescence and programmed cell death (Table I and Fig. 6), as they play crucial roles in regulating bone cell growth, influencing bone density and quality, affecting bone formation and repair processes as well as maintaining the balance of bone tissue (147).
Chromatin remodeling complexes and proliferation
Bone formation and remodeling during development are regulated by a network of transcription factors that facilitate the transition from proliferation to differentiation in osteoblastogenesis. Grandy et al (148) reported that the Ric-8B gene, a pro-proliferative factor, is negatively regulated during osteoblast differentiation by the transcription factor C/EBPb, which, along with BRG1 and BRM, inhibits Ric-8B promoter activity. This repression of Ric-8B expression is crucial as it influences osteoblast proliferation and differentiation, ultimately impacting bone development (148).
During bone development and remodeling, the proper proliferation of MSCs and ESCs is essential for maintaining stem cell pluripotency and regulating differentiation. However, uncontrolled proliferation can lead to the loss of stem cell function or abnormal differentiation. In this context, chromatin remodeling complexes play a critical role in maintaining the delicate balance between proliferation and differentiation. For instance, the loss of BAF250b in MSCs results in the upregulation of Bcl11b, which induces activin A and activates non-canonical activin signaling through the pERK pathway, impairing MSC quiescence and proliferation, thereby disrupting tissue homeostasis (149). Similarly, the loss of BRG1 initially leads to reduced proliferation, self-renewal and altered colony morphology in ESCs. Long-term depletion results in the complete loss of key ESC determinants, including OCT4, SOX2 and NANOG (150). Additionally, depletion of BAF155 in ESCs decreases proliferation and increases cell death, while forced expression of BAF170 reduces the competitive self-renewal ability of ESCs in immunocompromised mice and impairs teratoma formation (150,151). Furthermore, biallelic inactivation of BAF250b in ESCs leads to reduced proliferation, downregulation of pluripotency genes, increased expression of lineage-specific genes, including mesodermal differentiation markers, and cell cycle abnormalities, along with downregulation of the cell cycle regulator Cdc20 (152). Lastly, the nucleolar remodeling complex (NoRC), assembled by the histone variant H2A.X at ribosomal DNA (rDNA) promoters, recruits NoRC to rRNA genes, leading to the formation of a heterochromatin-like structure that downregulates rRNA transcription and ultimately reduces the proliferation rate of ESCs (153). Therefore, these findings highlight the intricate interplay between chromatin remodeling complexes and transcription factors in regulating proliferation, which is vital for proper bone development and tissue homeostasis.
Chromatin remodeling complexes and senescence
Cellular senescence, a pivotal aspect of biological aging, significantly influences bone development and related diseases (154). BRG1, serving as the ATPase subunit of the SWI/SNF chromatin remodeling complex, plays a crucial role in regulating cellular senescence in MSCs. Specifically, prior research has indicated that BRG1 is upregulated during the replicative senescence of MSCs, with forced BRG1 expression resulting in decreased telomerase activity and telomerase reverse transcriptase mRNA expression, coupled with the activation of RB- and p53-related pathways (155). Conversely, Alessio et al (156) reported that downregulation of BRG1 enhances senescence and heterochromatin formation via the activation of the RB2/P130 and p53 pathways, leading to reduced expression of stemness-related genes and compromised cellular physiology. Despite these seemingly inconsistent findings, the subsequent investigation by Squillaro et al (157) provided crucial insights, confirming that both forced upregulation and silencing of BRG1 in MSCs trigger cellular senescence. Specifically, in cells with silenced BRG1 expression, NANOG transcription is inhibited by RB1 and/or RB2-mediated DNA methyltransferase 1 (DNMT1) recruitment, causing methylation of the NANOG promoter, with BRM binding serving as a compensatory mechanism for the loss of BRG1. By contrast, in cells overexpressing BRG1, NANOG transcription is mainly suppressed through histone deacetylation, accompanied by reduced binding of DNMT1 and RB1 to the promoter, while histone deacetylase 1 and RB2 remain bound at E2F sites (157). Given these intricate interplays between BRG1 and cellular senescence in MSCs, it is evident that the role of chromatin remodeling complexes, particularly the SWI/SNF complex, in the senescence of osteoblasts, chondrocytes and osteoclasts remains an area ripe for further exploration.
Chromatin remodeling complexes and programmed cell death
Programmed cell death, including apoptosis, pyroptosis, autophagy and ferroptosis, plays a crucial role in the development, maintenance and repair of bone tissue (147). Apoptosis, a key form of programmed cell death, is essential for normal cell turnover, immune system function, embryonic development and the removal of damaged cells, ensuring the proper regulation of cellular processes (158). In bone development, chromatin remodeling complexes have been shown to regulate apoptosis, highlighting their critical role in maintaining cellular homeostasis. For example, BRG1 induces cell cycle arrest and apoptosis in MSCs, with p53 playing a central role in triggering programmed cell death (155). Similarly, SNF5 is vital for the survival of early embryonic cells, and its loss leads to apoptotic cell death and extensive DNA fragmentation (159). In the context of bone degeneration, BRD7 expression is decreased in severely degenerated NP tissues, and its knockdown promotes matrix degradation and apoptosis. Upregulation of BRD7 reverses these effects, enhancing matrix synthesis and reducing apoptosis through the PI3K and YAP1 pathways (160). These findings suggest that targeting the BRD7-PI3K-YAP1 axis, a chromatin remodeling pathway, could offer a potential therapeutic approach for treating intervertebral disc degeneration (IDD) by regulating apoptosis pathways.
Pyroptosis, an inflammatory form of programmed cell death typically triggered by inflammasomes and executed by gasdermin proteins in response to danger signals and pathogen infections, plays several crucial roles development, maintenance and repair, such as in osteogenic processes (161). Chai et al (162) discovered that tumor necrosis factor-α (TNF-α) induces pyroptosis in preosteoblastic MC3T3-E1 cells, negatively affecting osteogenic differentiation. Additionally, inhibiting Caspase-1-mediated pyroptosis promotes osteogenic differentiation, underscoring the complex relationship between these cell death pathways and bone development (163). A recent study has also shown that BRD9 contributes to bone degeneration through the regulation of inflammatory pathways via regulating pyroptosis (164). BRD9 expression increases as IDD progresses, and its inhibition reduces TNF-α-induced matrix breakdown, reactive oxygen species (ROS) production and pyroptosis in rat NP cells. BRD9 promotes IDD through the NADPH oxidase 1/ROS/NF-κB pathway, and in vivo inhibition of BRD9 helps mitigate IDD progression in a rat model (164). These findings suggest that targeting BRD9 and its chromatin remodeling functions may offer potential treatments for IDD, linking chromatin remodeling complexes and pyroptosis in bone development and degeneration.
Autophagy is a cellular process that facilitates the removal and recycling of damaged or unnecessary components to maintain bone homeostasis and function (165). A recent study has shown that BRG1 plays a crucial role in regulating bone development by upregulating pluripotency-related genes and activating pathways such as WNT/β-catenin signaling and autophagy, with inhibition of BRG1 leading to the opposite effect, suggesting an interconnected role of chromatin remodeling and autophagy in bone development and maintenance (166).
Role of chromatin remodeling complexes in the pathogenesis of bone-related diseases
The role of chromatin remodeling complexes in the pathogenesis of bone-related diseases is a critical area of study. Dysregulation of these complexes can impact bone cell differentiation, proliferation and apoptosis, resulting in a range of bone-related diseases, such as bone tumors, osteoporosis and OA. For example, low levels of BRD7 are associated with a poor prognosis in patients with osteosarcoma, while upregulation of BAF155 triggers OA (Table II). Understanding the mechanisms by which chromatin remodeling complexes contribute to these pathologies could reveal new therapeutic targets for treating bone-related diseases.
Chromatin remodeling complexes and bone tumors
Bone tumors refer to abnormal cellular proliferation occurring in the bone or surrounding tissues (such as cartilage and bone marrow) and can be classified as either benign or malignant (10,167). Although malignant bone tumors are rare [the annual incidence rate of osteosarcoma is 4-5 individuals per million (168)], the early symptoms are often subtle, leading to misdiagnosis as 'growing pains' or arthritis, which delays treatment (169-171). Common types of bone tumors include benign tumors, such as chondroma and giant cell tumor of the bone, the latter of which may undergo malignant transformation (172). Malignant bone tumors can be primary, including osteosarcoma (predominantly affecting adolescents), chondrosarcoma (more common in middle-aged and elderly individuals) and Ewing sarcoma (frequently seen in children and adolescents) (173,174). Alternatively, malignant bone tumors can be metastatic, originating from cancer types such as lung or breast cancer and subsequently spreading to the bone (175).
Chromatin remodeling complexes play a critical role in regulating tumor cell proliferation, survival and metastasis across various bone tumors. In Ewing sarcoma, for example, CHD4 depletion leads to increased DNA accessibility, which in turn increases DNA damage (176). Moreover, Ewing sarcoma is driven by the EWS-FLI1 fusion protein, which alters gene expression through the prion-like domains of EWS and ARID1A, facilitating the formation of biomolecular condensates crucial for tumor progression. ARID1A condensates localize to EWS/FLI1 target enhancers, driving long-range chromatin remodeling and the formation of functional hubs at oncogenic genes (177). Selvanathan et al (178) describe a novel feed-forward cycle in Ewing sarcoma, where EWS-FLI1 promotes the splicing of the ARID1A isoform protein variant ARID1A-L, driving tumor growth, while ARID1A-L, in turn, stabilizes EWS-FLI1, revealing potential targets for cancer-specific therapies. Additionally, in the presence of EWS-FLI1, multimerization at GGAA repeats and the prion-like domains of EWSR1 facilitate the recruitment of BAF complexes to tumor-specific enhancers, driving oncogenic gene expression (179). Autocrine neural EGFL like 2 (NELL2) signaling in Ewing sarcoma cells regulates BAF chromatin remodeling complexes and promotes cell proliferation by inhibiting Cdc42 and upregulating EWS-FLI1, with NELL2, CD133 and EWS-FLI1 positively regulating each other to enhance proliferative capacity (180). Similarly, in synovial sarcoma, the SS18-SSX fusion protein disrupts normal chromatin remodeling, leading to tumorigenesis (181). Additionally, ncBAF complexes uniquely localize to CTCF sites and promoters, acting as synthetic lethal targets in synovial sarcoma, with BRD9 subunit depletion effectively reducing cell proliferation (182). In chondrosarcoma, proline-rich polypeptide-1 inhibits chondrosarcoma cancer stem cell proliferation by targeting the SWI/SNF chromatin-remodeling complex, reducing the expression of key oncogenic players such as BRG, BAF170 and BRM (62). EMC is characterized by alterations in BAF47, although the precise mechanisms remain unclear (98,99). Osteosarcoma, the most prevalent primary bone sarcoma, is associated with reduced levels of BAF47 and BAF250a, suggesting their potential involvement in tumor progression (69,173,183-185). BRD7 also functions as a tumor suppressor in osteosarcoma and is degraded by the APC/C complex during the cell cycle. A BRD7 mutant resistant to APC/C degradation more effectively suppresses tumor growth, suggesting that targeting the APC/C-BRD7 pathway, along with APC/C inhibitors such as proTAME, could provide a novel therapeutic approach for osteosarcoma (126). Additionally, in response to doxorubicin treatment in U2OS cells, the INO80 complex regulates the removal of H2A.Z at the p21 promoter, facilitating p21 activation and contributing to the DNA damage response in osteosarcoma (186). Another study uncovered that pRB directly activates the osteoblast differentiation marker, Alpl, by recruiting the BRG1-containing SWI/SNF chromatin-remodeling complex, which displaces BRM-containing complexes, marking the onset of differentiation and highlighting the critical role of pRB in osteosarcoma susceptibility (187). Moreover, cohesin and PBAF play a crucial role in suppressing gene transcription near DNA double-strand breaks during interphase, and their absence results in extensive genome alterations, potentially driving genomic instability in osteosarcoma (188). The PHD domain of the KAP1 corepressor acts as an intramolecular E3 SUMO ligase, facilitating sumoylation of the adjacent bromodomain, which is crucial for KAP1-mediated gene silencing in osteosarcoma by recruiting SETDB1 and the CHD3/Mi2 NuRD complex, thereby enhancing histone methyltransferase activity (189). Similarly, poorly differentiated chordoma also exhibits BAF47 loss, implicating its role in tumor biology (69,183,185,190). In skull base chordomas, BAF180 mutations are linked to poor prognosis, underscoring the importance of chromatin remodeling complexes in determining tumor aggressiveness (191-193).
Given their central role in tumor progression, chromatin remodeling complexes present viable targets for therapeutic intervention. Trabectedin, for instance, has been shown to evict the SWI/SNF complex from chromatin in Ewing sarcoma, redistributing EWS-FLI1 and altering histone modifications to suppress tumor growth (194). The loss of CHD4 in Ewing sarcoma not only delays tumor growth and enhances overall survival but also, when combined with PARP inhibition using olaparib, significantly boosts the antitumor effects of genotoxic agents, positioning CHD4 as a promising therapeutic target (171). In synovial sarcoma, BRD9 inhibitors, such as FHD-609 (NCT04965753) (195) and CFT8634 (NCT05355753), have been developed to degrade BRD9, effectively impairing tumor proliferation (196,197). In osteosarcoma, BAZ1A and BRD7 have emerged as potential therapeutic targets, with BAZ1A influencing senescence and BRD7 stabilizing tumor suppression pathways (126,198). These findings suggest that targeting specific chromatin remodeling subunits could provide novel treatment strategies for sarcomas driven by epigenetic dysregulation.
Several chromatin remodeling components are frequently mutated or lost in bone and soft tissue sarcomas, highlighting their diagnostic and prognostic significance. In osteosarcoma, BRG1 mutations and the downregulation of BAF47 and BAF250a suggest a disrupted chromatin landscape (69,173,183-185). Similarly, synovial sarcoma is characterized by the SS18-SSX fusion, which affects SMARCB1 function (104,105). BAF47 loss is a hallmark of poorly differentiated chordoma and EMC, whereas BAF180 mutations in skull base chordomas are associated with poor prognosis (98,99,191-193). These genetic alterations could serve as valuable biomarkers for the early detection, risk stratification and treatment response monitoring, reinforcing the clinical relevance of chromatin remodeling complexes in sarcoma pathology.
Chromatin remodeling complexes and OA
OA, the most common joint disorder primarily affecting the aging population, is a leading musculoskeletal cause of impaired mobility, with the majority of individuals >65 years experiencing this condition (199). Wang et al (200) reported that interferon regulatory factor 1 (IRF1) regulates BAF155 expression, playing a crucial role in OA development, as knockdown of either IRF1 or BAF155 alleviates OA-like symptoms and inflammatory responses. Epigenetic modifications near the BAF155 promoter, mediated by GCN5 and SETD2, are key drivers of its expression, with upregulation of these enzymes worsening OA symptoms (200). Additionally, a study found that reduced levels of BRD7 activates the main immunodeficiency pathway. By contrast, increased levels of BRD7 may activate the ECM receptor interaction pathway and inhibit the primary immunodeficiency pathway, ultimately leading to ferroptosis in OA (201).
Chromatin remodeling complexes and bone-fat disorder
Bone marrow is the only tissue where bone and fat are adjacent and collaborate in various processes such as metabolic homeostasis, hemopoiesis and osteogenesis (202). Successful osteogenesis and adipogenesis are tightly regulated processes that must maintain a balance for proper skeletal and metabolic function. It has been reported that PPARγ serves as a differentiation switch between osteoblastogenesis and adipogenesis in the bone marrow, inhibiting the process of osteoblastogenesis (128). Depletion of CHD7 in MSCs and preosteoblasts disrupts this balance, leading to low bone mass and increased marrow adiposity through enhanced PPARγ signaling and trimethylated H3K4-mediated activation of lipogenic genes (127-129).
Chromatin remodeling complexes and AIS
AIS is defined by a lateral curvature of the spine (Cobb angle) of at least 10 degrees, occurring in the absence of any underlying congenital or neuromuscular abnormalities (203). The Cobb angle is calculated by drawing lines along the most tilted vertebrae at the curve's top and bottom and measuring the intersection (204). Studies have found that genetic variants of CHD7 play a vital role in AIS. According to a comparison of genotype and allele frequency between patients with AIS and healthy controls, rs121434341 of CHD7 is significantly associated with AIS susceptibility (205). Meanwhile, the rs1017861 polymorphism in the CHD7 gene is associated with susceptibility idiopathic scoliosis in Caucasian female patients, averaging 16.8±4.2 years old (range, 12.3-50.1). Additionally, the rs1017861 and rs4738813 polymorphisms are associated with higher curvature severity and progression rate (206). Lower expression of CHD7 has also been observed in patients with AIS compared with controls, which may be associated with the etiology of aberrant bone mass in AIS, but the pathogenetic mutation mechanism remains to be determined (205,207).
Chromatin remodeling complexes and osteoporosis
During senescence, the bone marrow has a reduced number of MSCs and an impaired balance between osteogenesis and osteoclastogenesis, leading to age-related osteoporosis (131,208,209). Therefore, the chromatin remodeling complex that contributes to osteogenesis and osteoclastogenesis may play a significant role in osteoporosis, such as BRM, BRD9 and CHD1 (14,64,210). BRM plays a crucial role in maintaining the balance of MSC lineage commitment between adipocytes and osteoblasts. BRM-null mice show a marked resistance to osteoporosis as the population of osteoblast progenitors is enhanced at the expense of increased adipocyte differentiation (64). Glucocorticoids, while effective for treating inflammatory and autoimmune disorders, cause bone loss by repressing osteogenic genes through the glucocorticoid receptor (GR), particularly involving the BRM. A study has revealed that BRM, unlike the essential ATPase BRG1, cooperates with the GR to repress genes such as Bglap, Per3 and FasL, disrupting osteoblast differentiation and promoting bone degradation (211). Moreover, BRD9 deficiency enhances osteoclast commitment and accelerates the development of osteoporosis (14). Postmenopausal bone loss, related to estrogen deficiency, is the predominant contributor to osteoporosis. Serum levels of CHD1 are elevated during postmenopausal osteoporosis (PMOP) and may play a role in pathogenesis. This elevation could potentially serve as an effective marker for diagnosing PMOP (210).
Conclusions and perspectives
Chromatin remodeling complexes are crucial for maintaining normal bone development and are implicated in various bone-related diseases. The present review provides a detailed examination of the diverse roles of these complexes, revealing their functions as regulators, promoters, inhibitors and stabilizers in bone biology. The present review emphasizes that understanding the specific mechanisms by which chromatin remodeling complexes influence bone homeostasis is essential for developing targeted therapies. For instance, certain subunits of these complexes may play critical roles in modulating gene expression during osteoblast differentiation, and disruptions in their function could lead to conditions such as osteoporosis or other metabolic bone diseases. Identifying these specific subunits and their interactions will be vital for designing interventions that can restore normal bone function.
However, the significant gap in research regarding the impact of aging on the functionality of chromatin remodeling complexes in bone health should be noted. The few studies available suggest that age-related changes in these complexes may contribute to impaired bone regeneration and increased fracture risk. This highlights the need for longitudinal studies that examine how the activity and composition of chromatin remodeling complexes evolve with age, and how these changes correlate with clinical outcomes in bone health. Understanding these dynamics will be crucial for developing strategies to mitigate the effects of aging on bone integrity.
Furthermore, it is important to consider the potential therapeutic implications of targeting chromatin remodeling complexes. Given their central role in regulating gene expression, pharmacological modulation of these complexes could provide a novel approach to treating bone-related diseases. For example, small molecules that enhance the activity of specific chromatin remodeling complexes may promote osteoblast differentiation and function, offering a new avenue for osteoporosis treatment. Conversely, small molecule inhibitors that target dysfunctional subunits of these complexes could help restore normal gene expression patterns, thereby addressing the underlying causes of metabolic bone diseases, such as using dBRD9 to treat synovial sarcoma and trabectedin for Ewing sarcoma (194,196). Controlled lineage selection in MSCs offers a promising strategy for cell engineering and tissue regeneration. For instance, CHD7-silenced MSCs show comprised osteogenic ability when cultured with a scaffold in vivo (84). Since BRM remains unchanged during normal osteogenesis, targeting it may enhance bone formation, and developing BRM inhibitors holds promise as a potential osteoporosis treatment (64). Targeting BRM, a key mediator of GR-induced repression in osteogenesis, may also offer a therapeutic strategy to counteract glucocorticoid-induced bone loss and potentially aid in reversing age-related osteoporosis by relieving repression on osteogenic genes (211). Furthermore, a study has shown that transfection of MSCs with small interfering RNA targeting INO80 results in impaired mineral deposition under osteogenic induction conditions, and mice implanted with INO80-silenced MSCs also exhibit reduced bone formation. This highlights the critical role of the INO80 complex in MSC osteogenic differentiation and its potential applications in clinical tissue engineering and osteoporosis treatment (131). BAF250a, another subunit, acts as a key link between EWs:Fli1 and the BAF complex; its capacity to form condensates is crucial for the dysregulated gene expression that fuels growth. As a result, the absence of condensate-competent BAF250a significantly obstructs tumor advancement, highlighting it as a possible therapeutic target. Nonetheless, the transient nature of these interactions complicates efforts to effectively target condensate formation, posing challenges in creating suitable inhibitors (177). In clinical trials, some new drugs are still under development and in the early phase such as FHD-609, a heterobifunctional degrader of BRD9 that treats advanced synovial sarcoma or SMARCB1-deficient tumors, which is in phase I study (195). Moreover, tazemetostat has demonstrated some efficacy in treating certain SMARCB1-deficient tumors in a subset of patients. However, it still faces limitations such as inconsistent efficacy, unclear indications and ambiguous dosing guidelines (212). Therefore, the potential therapeutic role of chromatin remodeling complexes in bone-related diseases needs further studies and research.
Additionally, through the continuous exploration of the assembly and 3D structures of chromatin remodeling complex subunits, there is potential for the discovery and development of specific chemical modulators that can optimize therapeutic outcomes. Understanding the structural nuances of these complexes will facilitate the design of targeted interventions that can more effectively influence their activity in bone biology. By leveraging structural biology techniques, such as X-ray crystallography and cryo-electron microscopy, researchers can gain insights into the conformational changes and interactions that govern the function of these complexes. For example, a potent and selective BRD9 bromodomain inhibitor series was developed based on a novel pyridinone-like scaffold, with crystallographic data guiding the optimization of its potency for BRD9 (213). Moreover, the integration of artificial intelligence in structural analysis presents a transformative opportunity for advancing our understanding of chromatin remodeling complexes. This approach can significantly accelerate the discovery of small molecules that specifically target the active or allosteric sites of chromatin remodeling complexes, enhancing their therapeutic efficacy.
In conclusion, while there has been progress in understanding the role of chromatin remodeling complexes in bone-related diseases, our knowledge remains limited and fragmented. We consider that utilizing advanced techniques, such as CRISPR-based gene editing and high-throughput sequencing, will allow for a more comprehensive exploration of these complexes and their interactions within the bone microenvironment. Additionally, integrating insights from systems biology could help elucidate the complex regulatory networks involving chromatin remodeling in bone biology. This deeper understanding could ultimately lead to innovative therapeutic strategies aimed at enhancing bone health and effectively treating degenerative bone conditions, thereby improving patient outcomes and quality of life.
Supplementary Data
Availability of data and materials
Not applicable.
Authors' contributions
WW prepared the first draft of the manuscript; YW, YN and CZ performed the literature search; WW and YC wrote and edited the manuscript; WW and QY drew the figures; WS and QY supervised and polished the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Abbreviations:
lncRNAs |
long non-coding RNAs |
SWI/SNF |
switch/sucrose-non-fermenting |
ISWI |
imitation switch |
CHD |
chromodomain-helicase-DNA binding |
INO80/SWR |
inositol requiring 80/SWi2/snf2-related 1 |
EWS-FLI1 |
Ewing sarcoma-friend leukemia integration 1 |
EWSR1 |
Ewing sarcoma RNA binding protein 1 |
hESCs |
human embryonic stem cells |
FGFR3 |
fibroblast growth factor receptor 3 |
BMP |
bone morphogenic protein |
MSCs |
mesenchymal stem cells |
PBAF |
polybromo BAF |
TSS |
transcriptional start site |
PPAR |
peroxisome proliferator-activated receptor |
IDD |
intervertebral disc degeneration |
NP |
nucleus pulposus |
OA |
osteoarthritis |
AIS |
adolescent idiopathic scoliosis |
PMOP |
postmenopausal osteoporosis |
Acknowledgments
Not applicable.
Funding
This research was supported by Sichuan Science and Technology Program (grant nos. 2022YFS0609 and 2022NSFSC1368), Scientific Research Foundation of Southwest Medical University (grant no. 2021ZKMS009) and Shenzhen Science and Technology Projects (grant nos. JCYJ20210324103604013, JSGG20220831110400001 and KCXFZ20230731093059012).
References
Florencio-Silva R, Sasso GR, Sasso-Cerri E, Simões MJ and Cerri PS: Biology of bone tissue: Structure, function, and factors that influence bone cells. Biomed Res Int. 2015:4217462015. View Article : Google Scholar : PubMed/NCBI | |
Lee HR, Yang SJ, Choi HK, Kim JA and Oh IH: The chromatin remodeling complex CHD1 regulates the primitive state of mesenchymal stromal cells to control their stem cell supporting activity. Stem Cells Dev. 30:363–373. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schini M, Vilaca T, Gossiel F, Salam S and Eastell R: Bone turnover markers: Basic biology to clinical applications. Endocr Rev. 44:417–473. 2023. View Article : Google Scholar : | |
Hadjidakis DJ and Androulakis II: Bone remodeling. Ann N Y Acad Sci. 1092:385–396. 2006. View Article : Google Scholar | |
Liang J, Yi Q, Liu Y, Li J, Yang Z and Sun W and Sun W: Recent advances of m6A methylation in skeletal system disease. J Transl Med. 22:1532024. View Article : Google Scholar : PubMed/NCBI | |
Lane NE: Epidemiology, etiology, and diagnosis of osteoporosis. Am J Obstet Gynecol. 194(2 Suppl): S3–S11. 2006. View Article : Google Scholar : PubMed/NCBI | |
Stark Z and Savarirayan R: Osteopetrosis. Orphanet J Rare Dis. 4:52009. View Article : Google Scholar : PubMed/NCBI | |
Zheng J, Yao Z, Xue L, Wang D and Tan Z: The role of immune cells in modulating chronic inflammation and osteonecrosis. Front Immunol. 13:10642452022. View Article : Google Scholar : PubMed/NCBI | |
Glyn-Jones S, Palmer AJ, Agricola R, Price AJ, Vincent TL, Weinans H and Carr AJ: Osteoarthritis. Lancet. 386:376–387. 2015. View Article : Google Scholar : PubMed/NCBI | |
Choi JH and Ro JY: The 2020 WHO classification of tumors of bone: An updated review. Adv Anat Pathol. 28:119–138. 2021. View Article : Google Scholar : PubMed/NCBI | |
Park-Min KH: Epigenetic regulation of bone cells. Connect Tissue Res. 58:76–89. 2017. View Article : Google Scholar : | |
Zhang Y, Wang Q, Xue H, Guo Y, Wei S, Li F, Gong L, Pan W and Jiang P: Epigenetic regulation of autophagy in bone metabolism. Function (Oxf). 5:zqae0042024. View Article : Google Scholar : PubMed/NCBI | |
Sikora M, Marycz K and Smieszek A: Small and Long Non-coding RNAs as functional regulators of bone homeostasis, acting alone or cooperatively. Mol Ther Nucleic Acids. 21:792–803. 2020. View Article : Google Scholar : PubMed/NCBI | |
Du J, Liu Y, Wu X, Sun J, Shi J, Zhang H, Zheng A, Zhou M and Jiang X: BRD9-mediated chromatin remodeling suppresses osteoclastogenesis through negative feedback mechanism. Nat Commun. 14:14132023. View Article : Google Scholar : PubMed/NCBI | |
Busby T, Chen Y, Godfrey TC, Rehan M, Wildman BJ, Smith CM and Hassan Q: Baf45a mediated chromatin remodeling promotes transcriptional activation for osteogenesis and odontogenesis. Front Endocrinol (Lausanne). 12:7633922022. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Sun H, Huang F, Chen Y, Ding X, Zhou C, Wu Y, Zhang Q, Ma X, Wang J, et al: The chromatin remodeling factor Arid1a cooperates with Jun/Fos to promote osteoclastogenesis by epigenetically upregulating Siglec15 expression. J Bone Miner Res. 39:775–790. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wei W, Tang X, Jiang N, Ni C, He H, Sun S, Yu M, Yu C, Qiu M, Yan D, et al: Chromatin remodeler Znhit1 controls bone morphogenetic protein signaling in embryonic lung tissue branching. J Biol Chem. 298:1024902022. View Article : Google Scholar : PubMed/NCBI | |
Xue Y, Morris JL, Yang K, Fu Z, Zhu X, Johnson F, Meehan B, Witkowski L, Yasmeen A, Golenar T, et al: SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca(2+) flux to mitochondria. Nat Commun. 12:54042021. View Article : Google Scholar : PubMed/NCBI | |
Bosch PJ, Fuller LC and Weiner JA: A critical role for the nuclear protein Akirin2 in the formation of mammalian muscle in vivo. Genesis. 57:e232862019. View Article : Google Scholar : PubMed/NCBI | |
Gamarra N and Narlikar GJ; Collaboration through chromatin: motors of transcription and chromatin structure. J Mol Biol. 433:1668762021. View Article : Google Scholar | |
El Hadidy N and Uversky VN: Intrinsic disorder of the BAF complex: Roles in chromatin remodeling and disease development. Int J Mol Sci. 20:52602019. View Article : Google Scholar : PubMed/NCBI | |
Clapier CR and Cairns BR: The biology of chromatin remodeling complexes. Annu Rev Biochem. 78:273–304. 2009. View Article : Google Scholar : PubMed/NCBI | |
Fröb F and Wegner M: The role of chromatin remodeling complexes in Schwann cell development. Glia. 68:1596–1603. 2020. View Article : Google Scholar | |
Han Y, Reyes AA, Malik S and He Y: Cryo-EM structure of SWI/SNF complex bound to a nucleosome. Nature. 579:452–455. 2020. View Article : Google Scholar : PubMed/NCBI | |
Vary JC Jr, Gangaraju VK, Qin J, Landel CC, Kooperberg C, Bartholomew B and Tsukiyama T: Yeast Isw1p forms two separable complexes in vivo. Mol Cell Biol. 23:80–91. 2003. View Article : Google Scholar | |
Reyes AA, Marcum RD and He Y: Structure and function of chromatin remodelers. J Mol Biol. 433:1669292021. View Article : Google Scholar : PubMed/NCBI | |
Nacev BA, Jones KB, Intlekofer AM, Yu JSE, Allis CD, Tap WD, Ladanyi M and Nielsen TO: The epigenomics of sarcoma. Nat Rev Cancer. 20:608–623. 2020. View Article : Google Scholar : PubMed/NCBI | |
Du W, Guo D and Du W: ATP-Dependent chromatin remodeling complex in the lineage specification of mesenchymal stem cells. Stem Cells Int. 2020:88397032020. View Article : Google Scholar : PubMed/NCBI | |
Wojcik J and Cooper K: Epigenetic alterations in bone and soft tissue tumors. Adv Anat Pathol. 24:362–371. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chakraborty S, Sinha S and Sengupta A: Emerging trends in chromatin remodeler plasticity in mesenchymal stromal cell function. FASEB J. 35:e212342021. View Article : Google Scholar | |
Kouzarides T: Chromatin modifications and their function. Cell. 128:693–705. 2007. View Article : Google Scholar : PubMed/NCBI | |
Clapier CR, Iwasa J, Cairns BR and Peterson CL: Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat Rev Mol Cell Biol. 18:407–422. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tyagi M, Imam N, Verma K and Patel AK: Chromatin remodelers: We are the drivers! Nucleus. 7:388–404. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Gong H, Wang P, Zhu Y, Peng H, Cui Y, Li H, Liu J and Wang Z: The emerging role of ISWI chromatin remodeling complexes in cancer. J Exp Clin Cancer Res. 40:3462021. View Article : Google Scholar : PubMed/NCBI | |
Centore RC, Sandoval GJ, Soares LMM, Kadoch C and Chan HM: Mammalian SWI/SNF chromatin remodeling complexes: Emerging mechanisms and therapeutic strategies. Trends Genet. 36:936–950. 2020. View Article : Google Scholar : PubMed/NCBI | |
Willhoft O and Wigley DB: INO80 and SWR1 complexes: The non-identical twins of chromatin remodelling. Curr Opin Struct Biol. 61:50–58. 2020. View Article : Google Scholar : | |
Pulice JL and Kadoch C: Composition and function of mammalian SWI/SNF chromatin remodeling complexes in human disease. Cold Spring Harb Symp Quant Biol. 81:53–60. 2016. View Article : Google Scholar | |
Judd J, Duarte FM and Lis JT: Pioneer-like factor GAF cooperates with PBAP (SWI/SNF) and NURF (ISWI) to regulate transcription. Genes Dev. 35:147–156. 2021. View Article : Google Scholar : | |
Poli J, Gasser SM and Papamichos-Chronakis M: The INO80 remodeller in transcription, replication and repair. Philos Trans R Soc Lond B Biol Sci. 372:201602902017. View Article : Google Scholar : PubMed/NCBI | |
Trujillo JT, Long J, Aboelnour E, Ogas J and Wisecaver JH: CHD Chromatin Remodeling Protein Diversification Yields Novel Clades and Domains Absent in Classic Model Organisms. Genome Biol Evol. 14:evac0662022. View Article : Google Scholar : PubMed/NCBI | |
Iyer J, Gentry LK, Bergwell M, Smith A, Guagliardo S, Kropp PA, Sankaralingam P, Liu Y, Spooner E, Bowerman B and O'Connell KF: The chromatin remodeling protein CHD-1 and the EFL-1/DPL-1 transcription factor cooperatively down regulate CDK-2 to control SAS-6 levels and centriole number. PLoS Genet. 18:e10097992022. View Article : Google Scholar : PubMed/NCBI | |
Ebbert R, Birkmann A and Schüller HJ: The product of the SNF2/SWI2 paralogue INO80 of Saccharomyces cerevisiae required for efficient expression of various yeast structural genes is part of a high-molecular-weight protein complex. Mol Microbiol. 32:741–751. 1999. View Article : Google Scholar : PubMed/NCBI | |
van Attikum H, Fritsch O and Gasser SM: Distinct roles for SWR1 and INO80 chromatin remodeling complexes at chromosomal double-strand breaks. EMBO J. 26:4113–4125. 2007. View Article : Google Scholar : PubMed/NCBI | |
Conaway RC and Conaway JW: The INO80 chromatin remodeling complex in transcription, replication and repair. Trends Biochem Sci. 34:71–77. 2009. View Article : Google Scholar | |
Saha A, Wittmeyer J and Cairns BR: Mechanisms for nucleosome movement by ATP-dependent chromatin remodeling complexes. Results Probl Cell Differ. 41:127–148. 2006. View Article : Google Scholar : PubMed/NCBI | |
Barisic D, Stadler MB, Iurlaro M and Schübeler D: Mammalian ISWI and SWI/SNF selectively mediate binding of distinct transcription factors. Nature. 569:136–140. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al: The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 16:413–425. 2015. View Article : Google Scholar : PubMed/NCBI | |
Tang Y, Wang J, Lian Y, Fan C, Zhang P, Wu Y, Li X, Xiong F, Li X, Li G, et al: Linking long non-coding RNAs and SWI/SNF complexes to chromatin remodeling in cancer. Mol Cancer. 16:422017. View Article : Google Scholar : PubMed/NCBI | |
Patty BJ and Hainer SJ: Non-Coding RNAs and nucleosome remodeling complexes: An intricate regulatory relationship. Biology (Basel). 9:2132020.PubMed/NCBI | |
Han P, Li W, Lin CH, Yang J, Shang C, Nuernberg ST, Jin KK, Xu W, Lin CY, Lin CJ, et al: A long noncoding RNA protects the heart from pathological hypertrophy. Nature. 514:102–106. 2014. View Article : Google Scholar : PubMed/NCBI | |
Atala A: Re: The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. J Urol. 192:6132014. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, Mameri A, Cattoglio C, Lachance C, Florez Ariza AJ, Luo J, Humbert J, Sudarshan D, Banerjea A, Galloy M, et al: Structural insights into the human NuA4/TIP60 acetyltransferase and chromatin remodeling complex. Science. 385:eadl58162024. View Article : Google Scholar : PubMed/NCBI | |
Tallant C, Valentini E, Fedorov O, Overvoorde L, Ferguson FM, Filippakopoulos P, Svergun DI, Knapp S and Ciulli A: Molecular basis of histone tail recognition by human TIP5 PHD finger and bromodomain of the chromatin remodeling complex NoRC. Structure. 23:80–92. 2015. View Article : Google Scholar : | |
Charles GM, Chen C, Shih SC, Collins SR, Beltrao P, Zhang X, Sharma T, Tan S, Burlingame AL, Krogan NJ, et al: Site-specific acetylation mark on an essential chromatin-remodeling complex promotes resistance to replication stress. Proc Natl Acad Sci USA. 108:10620–10625. 2011. View Article : Google Scholar : PubMed/NCBI | |
Sala A, La Rocca G, Burgio G, Kotova E, Di Gesù D, Collesano M, Ingrassia AM, Tulin AV and Corona DF: The nucleosome-remodeling ATPase ISWI is regulated by poly-ADP-ribosylation. PLoS Biol. 6:e2522008. View Article : Google Scholar : PubMed/NCBI | |
Bure IV and Nemtsova MV: Mutual regulation of ncRNAs and chromatin remodeling complexes in normal and pathological conditions. Int J Mol Sci. 24:78482023. View Article : Google Scholar : PubMed/NCBI | |
Young DW, Pratap J, Javed A, Weiner B, Ohkawa Y, van Wijnen A, Montecino M, Stein GS, Stein JL, Imbalzano AN and Lian JB: SWI/SNF chromatin remodeling complex is obligatory for BMP2-induced, Runx2-dependent skeletal gene expression that controls osteoblast differentiation. J Cell Biochem. 94:720–730. 2005. View Article : Google Scholar | |
Kuhn NZ and Tuan RS: Regulation of stemness and stem cell niche of mesenchymal stem cells: Implications in tumorigenesis and metastasis. J Cell Physiol. 222:268–277. 2010. View Article : Google Scholar | |
Im GI and Shin KJ: Epigenetic approaches to regeneration of bone and cartilage from stem cells. Expert Opin Biol Ther. 15:181–193. 2015. View Article : Google Scholar | |
Flowers S, Nagl NG Jr, Beck GR Jr and Moran E: Antagonistic roles for BRM and BRG1 SWI/SNF complexes in differentiation. J Biol Chem. 284:10067–10075. 2009. View Article : Google Scholar : PubMed/NCBI | |
Xu Y, Zhang J and Chen X: The activity of p53 is differentially regulated by Brm- and Brg1-containing SWI/SNF chromatin remodeling complexes. J Biol Chem. 282:37429–37435. 2007. View Article : Google Scholar : PubMed/NCBI | |
Moran A, Hoyt A, Sedani A, Granger C, Saigh S, Blonska M, Zhao-Ju L, Conway SA, Pretell J, Brown J and Galoian K: Proline-rich polypeptide-1 decreases cancer stem cell population by targeting BAFF chromatin-remodeling complexes in human chondrosarcoma JJ012 cells. Oncol Rep. 44:393–403. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kidder BL, Palmer S and Knott JG: SWI/SNF-Brg1 regulates self-renewal and occupies core pluripotency-related genes in embryonic stem cells. Stem Cells. 27:317–328. 2009. View Article : Google Scholar | |
Nguyen KH, Xu F, Flowers S, Williams EA, Fritton JC and Moran E: SWI/SNF-mediated lineage determination in mesenchymal stem cells confers resistance to osteoporosis. Stem Cells. 33:3028–3038. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kitagawa T, Kobayashi D, Baron B, Okita H, Miyamoto T, Takai R, Paudel D, Ohta T, Asaoka Y, Tokunaga M, et al: AT-hook DNA-binding motif-containing protein one knockdown downregulates EWS-FLI1 transcriptional activity in Ewing's sarcoma cells. PLoS One. 17:e02690772022. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Lee RS, Alver BH, Haswell JR, Wang S, Mieczkowski J, Drier Y, Gillespie SM, Archer TC, Wu JN, et al: SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet. 49:289–295. 2017. View Article : Google Scholar : | |
Nakayama RT, Pulice JL, Valencia AM, McBride MJ, McKenzie ZM, Gillespie MA, Ku WL, Teng M, Cui K, Williams RT, et al: SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters. Nat Genet. 49:1613–1623. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Wang X, Li J, Shi R and Ye Y: BAF Complex in embryonic stem cells and early embryonic development. Stem Cells Int. 2021:66688662021. View Article : Google Scholar : PubMed/NCBI | |
Antonelli M, Raso A, Mascelli S, Gessi M, Nozza P, Coli A, Gardiman MP, Arcella A, Massimino M, Buttarelli FR and Giangaspero F: SMARCB1/INI1 involvement in pediatric chordoma: A mutational and immunohistochemical analysis. Am J Surg Pathol. 41:56–61. 2017. View Article : Google Scholar | |
Zhang X, Li B, Li W, Ma L, Zheng D, Li L, Yang W, Chu M, Chen W, Mailman RB, et al: Transcriptional repression by the BRG1-SWI/SNF complex affects the pluripotency of human embryonic stem cells. Stem Cell Reports. 3:460–474. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gatchalian J, Malik S, Ho J, Lee DS, Kelso TWR, Shokhirev MN, Dixon JR and Hargreaves DC: A non-canonical BRD9-containing BAF chromatin remodeling complex regulates naive pluripotency in mouse embryonic stem cells. Nat Commun. 9:51392018. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Song C, Ye Y, Gu Y, Li X, Chen P, Leng D, Xiao J, Wu H, Xie S, et al: BRD9-mediated control of the TGF-β/Activin/Nodal pathway regulates self-renewal and differentiation of human embryonic stem cells and progression of cancer cells. Nucleic Acids Res. 51:11634–11651. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sevinç K, Sevinç GG, Cavga AD, Philpott M, Kelekçi S, Can H, Cribbs AP, Yıldız AB, Yılmaz A, Ayar ES, et al: BRD9-containing non-canonical BAF complex maintains somatic cell transcriptome and acts as a barrier to human reprogramming. Stem Cell Reports. 17:2629–2642. 2022. View Article : Google Scholar : PubMed/NCBI | |
Gaspar-Maia A, Alajem A, Polesso F, Sridharan R, Mason MJ, Heidersbach A, Ramalho-Santos J, McManus MT, Plath K, Meshorer E and Ramalho-Santos M: Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature. 460:863–868. 2009. View Article : Google Scholar : PubMed/NCBI | |
Baumgart SJ, Najafova Z, Hossan T, Xie W, Nagarajan S, Kari V, Ditzel N, Kassem M and Johnsen SA: CHD1 regulates cell fate determination by activation of differentiation-induced genes. Nucleic Acids Res. 45:7722–7735. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bulut-Karslioglu A, Jin H, Kim YK, Cho B, Guzman-Ayala M, Williamson AJK, Hejna M, Stötzel M, Whetton AD, Song JS and Ramalho-Santos M: Chd1 protects genome integrity at promoters to sustain hypertranscription in embryonic stem cells. Nat Commun. 12:48592021. View Article : Google Scholar : PubMed/NCBI | |
Caplan AI: Mesenchymal stem cells. J Orthop Res. 9:641–650. 1991. View Article : Google Scholar : PubMed/NCBI | |
Benayahu D, Shacham N and Shur I: Insights on the functional role of chromatin remodelers in osteogenic cells. Crit Rev Eukaryot Gene Expr. 17:103–113. 2007. View Article : Google Scholar : PubMed/NCBI | |
Suzuki S, Nozawa Y, Tsukamoto S, Kaneko T, Manabe I, Imai H and Minami N: CHD1 acts via the Hmgpi pathway to regulate mouse early embryogenesis. Development. 142:2375–2384. 2015.PubMed/NCBI | |
Liu C, Kang N, Guo Y and Gong P: Advances in chromodomain helicase DNA-binding (CHD) proteins regulating stem cell differentiation and human diseases. Front Cell Dev Biol. 9:7102032021. View Article : Google Scholar : PubMed/NCBI | |
Wan M, Liang J, Xiong Y, Shi F, Zhang Y, Lu W, He Q, Yang D, Chen R, Liu D, et al: The trithorax group protein Ash2l is essential for pluripotency and maintaining open chromatin in embryonic stem cells. J Biol Chem. 288:5039–5048. 2013. View Article : Google Scholar : | |
Yang P, Oldfield A, Kim T, Yang A, Yang JYH and Ho JWK: Integrative analysis identifies co-dependent gene expression regulation of BRG1 and CHD7 at distal regulatory sites in embryonic stem cells. Bioinformatics. 33:1916–1920. 2017. View Article : Google Scholar : PubMed/NCBI | |
Malla S, Martinez-Gamero C, Kumari K, Achour C, Mermelekas G, Martinez-Delgado D, Coego A, Guallar D, Roman AC and Aguilo F: Cooperative role of LSD1 and CHD7 in regulating differentiation of mouse embryonic stem cells. Sci Rep. 14:284952024. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Wang M, Chen D, Wang J and Kang N: Chromatin remodeling enzyme CHD7 is necessary for osteogenesis of human mesenchymal stem cells. Biochem Biophys Res Commun. 478:1588–1593. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yoo H, La H, Lee EJ, Choi HJ, Oh J, Thang NX and Hong K: ATP-dependent chromatin remodeler CHD9 controls the proliferation of embryonic stem cells in a cell culture condition-dependent manner. Biology (Basel). 9:4282020.PubMed/NCBI | |
Salomon-Kent R, Marom R, John S, Dundr M, Schiltz LR, Gutierrez J, Workman J, Benayahu D and Hager GL: New face for chromatin-related mesenchymal modulator: n-CHD9 localizes to nucleoli and interacts with ribosomal genes. J Cell Physiol. 230:2270–2280. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Du Y, Ward JM, Shimbo T, Lackford B, Zheng X, Miao YL, Zhou B, Han L, Fargo DC, et al: INO80 facilitates pluripotency gene activation in embryonic stem cell self-renewal, reprogramming, and blastocyst development. Cell Stem Cell. 14:575–591. 2014. View Article : Google Scholar : PubMed/NCBI | |
Furumatsu T and Ozaki T: Epigenetic regulation in chondrogenesis. Acta Med Okayama. 64:155–161. 2010.PubMed/NCBI | |
Berendsen AD and Olsen BR: Bone development. Bone. 80:14–18. 2015. View Article : Google Scholar : PubMed/NCBI | |
Mashtalir N, D'Avino AR, Michel BC, Luo J, Pan J, Otto JE, Zullow HJ, McKenzie ZM, Kubiak RL, St Pierre R, et al: Modular organization and assembly of SWI/SNF family chromatin remodeling complexes. Cell. 175:1272–1288.e20. 2018. View Article : Google Scholar : PubMed/NCBI | |
You S, Zhang Y, Xu J, Qian H, Wu S, Wu B, Lu S, Sun Y and Zhang N: The role of BRG1 in antioxidant and redox signaling. Oxid Med Cell Longev. 2020:60956732020. View Article : Google Scholar : PubMed/NCBI | |
Sun F, Chen Q, Yang S, Pan Q, Ma J, Wan Y, Chang CH and Hong A: Remodeling of chromatin structure within the promoter is important for bmp-2-induced fgfr3 expression. Nucleic Acids Res. 37:3897–3911. 2009. View Article : Google Scholar : PubMed/NCBI | |
Sacitharan PK, Lwin S, Gharios GB and Edwards JR: Spermidine restores dysregulated autophagy and polyamine synthesis in aged and osteoarthritic chondrocytes via EP300. Exp Mol Med. 50:1–10. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chen Z, Lin CX, Song B, Li CC, Qiu JX, Li SX, Lin SP, Luo WQ, Fu Y, Fang GB, et al: Spermidine activates RIP1 deubiquitination to inhibit TNF-α-induced NF-κB/p65 signaling pathway in osteoarthritis. Cell Death Dis. 11:5032020. View Article : Google Scholar | |
Guo X, Feng X, Yang Y, Zhang H and Bai L: Spermidine attenuates chondrocyte inflammation and cellular pyroptosis through the AhR/NF-κB axis and the NLRP3/caspase-1/GSDMD pathway. Front Immunol. 15:14627772024. View Article : Google Scholar | |
Mao X, Yan B, Chen H, Lai P and Ma J: BRG1 mediates protective ability of spermidine to ameliorate osteoarthritic cartilage by Nrf2/KEAP1 and STAT3 signaling pathway. Int Immunopharmacol. 122:1105932023. View Article : Google Scholar : PubMed/NCBI | |
Wei S, Pei J, von Mehren M, Abraham JA, Patchefsky AS and Cooper HS: SMARCA2-NR4A3 is a novel fusion gene of extraskeletal myxoid chondrosarcoma identified by RNA next-generation sequencing. Genes Chromosomes Cancer. 60:709–712. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schaefer IM and Hornick JL: SWI/SNF complex-deficient soft tissue neoplasms: An update. Semin Diagn Pathol. 38:222–231. 2021. View Article : Google Scholar : | |
Rekhi B and Vogel U: Utility of characteristic 'Weak to Absent' INI1/SMARCB1/BAF47 expression in diagnosis of synovial sarcomas. Apmis. 123:618–628. 2015. View Article : Google Scholar : PubMed/NCBI | |
Fanburg-Smith JC, Auerbach A, Marwaha JS, Wang Z, Santi M, Judkins AR and Rushing EJ: Immunoprofile of mesenchymal chondrosarcoma: Aberrant desmin and EMA expression, retention of INI1, and negative estrogen receptor in 22 female-predominant central nervous system and musculoskeletal cases. Ann Diagn Pathol. 14:8–14. 2010. View Article : Google Scholar : PubMed/NCBI | |
Kohashi K, Oda Y, Yamamoto H, Tamiya S, Matono H, Iwamoto Y, Taguchi T and Tsuneyoshi M: Reduced expression of SMARCB1/INI1 protein in synovial sarcoma. Mod Pathol. 23:981–990. 2010. View Article : Google Scholar : PubMed/NCBI | |
Shain AH and Pollack JR: The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS One. 8:e551192013. View Article : Google Scholar : PubMed/NCBI | |
Kadoch C, Williams RT, Calarco JP, Miller EL, Weber CM, Braun SM, Pulice JL, Chory EJ and Crabtree GR: Dynamics of BAF-Polycomb complex opposition on heterochromatin in normal and oncogenic states. Nat Genet. 49:213–222. 2017. View Article : Google Scholar | |
Cheng Y, Shen Z, Gao Y, Chen F, Xu H, Mo Q, Chu X, Peng CL, McKenzie TT, Palacios BE, et al: Phase transition and remodeling complex assembly are important for SS18-SSX oncogenic activity in synovial sarcomas. Nat Commun. 13:27242022. View Article : Google Scholar : PubMed/NCBI | |
McBride MJ, Pulice JL, Beird HC, Ingram DR, D'Avino AR, Shern JF, Charville GW, Hornick JL, Nakayama RT, Garcia-Rivera EM, et al: The SS18-SSX fusion oncoprotein hijacks BAF complex targeting and function to drive synovial sarcoma. Cancer Cell. 33:1128–1141.e7. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang N, Zhang Y, Chen Y, Qian H, Wu B, Lu S, You S, Xu W, Zou Y, Huang X, et al: BAF155 promotes cardiac hypertrophy and fibrosis through inhibition of WWP2-mediated PARP1 ubiquitination. Cell Discov. 9:462023. View Article : Google Scholar : PubMed/NCBI | |
Jeon S and Seong RH: Anteroposterior limb skeletal patterning requires the bifunctional action of SWI/SNF chromatin remodeling complex in hedgehog pathway. PLoS Genet. 12:e10059152016. View Article : Google Scholar : PubMed/NCBI | |
Ju C, Liu R, Zhang YW, Zhang Y, Zhou R, Sun J, Lv XB and Zhang Z: Mesenchymal stem cell-associated lncRNA in osteogenic differentiation. Biomed Pharmacother. 115:1089122019. View Article : Google Scholar : PubMed/NCBI | |
Peng Y, Jiang H and Zuo HD: Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells. 15:548–560. 2023. View Article : Google Scholar : PubMed/NCBI | |
Toosi S and Behravan J: Osteogenesis and bone remodeling: A focus on growth factors and bioactive peptides. Biofactors. 46:326–340. 2020. View Article : Google Scholar | |
Gou Y, Huang Y, Luo W, Li Y, Zhao P, Zhong J, Dong X, Guo M, Li A, Hao A, et al: Adipose-derived mesenchymal stem cells (MSCs) are a superior cell source for bone tissue engineering. Bioact Mater. 34:51–63. 2023. | |
Sinha S, Biswas M, Chatterjee SS, Kumar S and Sengupta A: Pbrm1 steers mesenchymal stromal cell osteolineage differentiation by integrating PBAF-dependent chromatin remodeling and BMP/TGF-β signaling. Cell Rep. 31:1075702020. View Article : Google Scholar | |
Mardinian K, Adashek JJ, Botta GP, Kato S and Kurzrock R: SMARCA4: Implications of an altered chromatin-remodeling gene for cancer development and therapy. Mol Cancer Ther. 20:2341–2351. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hojo H, Saito T, He X, Guo Q, Onodera S, Azuma T, Koebis M, Nakao K, Aiba A, Seki M, et al: Runx2 regulates chromatin accessibility to direct the osteoblast program at neonatal stages. Cell Rep. 40:1113152022. View Article : Google Scholar : PubMed/NCBI | |
Nagl NG Jr, Patsialou A, Haines DS, Dallas PB, Beck GR Jr and Moran E: The p270 (ARID1A/SMARCF1) subunit of mammalian SWI/SNF-related complexes is essential for normal cell cycle arrest. Cancer Res. 65:9236–9244. 2005. View Article : Google Scholar : PubMed/NCBI | |
Bailey S, Karsenty G, Gundberg C and Vashishth D: Osteocalcin and osteopontin influence bone morphology and mechanical properties. Ann N Y Acad Sci. 1409:79–84. 2017. View Article : Google Scholar : PubMed/NCBI | |
Komori T: Functions of osteocalcin in bone, pancreas, testis, and muscle. Int J Mol Sci. 21:75132020. View Article : Google Scholar : PubMed/NCBI | |
Villagra A, Cruzat F, Carvallo L, Paredes R, Olate J, van Wijnen AJ, Stein GS, Lian JB, Stein JL, Imbalzano AN and Montecino M: Chromatin remodeling and transcriptional activity of the bone-specific osteocalcin gene require CCAAT/enhancer-binding protein beta-dependent recruitment of SWI/SNF activity. J Biol Chem. 281:22695–22706. 2006. View Article : Google Scholar : PubMed/NCBI | |
Flowers S, Patel PJ, Gleicher S, Amer K, Himelman E, Goel S and Moran E: p107-Dependent recruitment of SWI/SNF to the alkaline phosphatase promoter during osteoblast differentiation. Bone. 69:47–54. 2014. View Article : Google Scholar : PubMed/NCBI | |
Reyes JC, Barra J, Muchardt C, Camus A, Babinet C and Yaniv M: Altered control of cellular proliferation in the absence of mammalian brahma (SNF2alpha). EMBO J. 17:6979–6991. 1998. View Article : Google Scholar : PubMed/NCBI | |
Middeljans E, Wan X, Jansen PW, Sharma V, Stunnenberg HG and Logie C: SS18 together with animal-specific factors defines human BAF-type SWI/SNF complexes. PLoS One. 7:e338342012. View Article : Google Scholar : PubMed/NCBI | |
Godfrey TC, Wildman BJ, Javed A, Lengner CJ and Hassan MQ: Epigenetic remodeling and modification to preserve skeletogenesis in vivo. Connect Tissue Res. 59(supp1): S52–S54. 2018. View Article : Google Scholar | |
Hasenfratz M, Mellert K, Marienfeld R, von Baer A, Schultheiss M, Roitman PD, Aponte-Tinao LA, Lehner B, Möller P, Mechtersheimer G and Barth TFE: Profiling of three H3F3A-mutated and denosumab-treated giant cell tumors of bone points to diverging pathways during progression and malignant transformation. Sci Rep. 11:57092021. View Article : Google Scholar : PubMed/NCBI | |
Xu F, Flowers S and Moran E: Essential role of ARID2 protein-containing SWI/SNF complex in tissue-specific gene expression. J Biol Chem. 287:5033–5041. 2012. View Article : Google Scholar : | |
Wilsker D, Patsialou A, Dallas PB and Moran E: ARID proteins: A diverse family of DNA binding proteins implicated in the control of cell growth, differentiation, and development. Cell Growth Differ. 13:95–106. 2002.PubMed/NCBI | |
Hu K, Liao D, Wu W, Han AJ, Shi HJ, Wang F, Wang X, Zhong L, Duan T, Wu Y, et al: Targeting the anaphase-promoting complex/cyclosome (APC/C)-bromodomain containing 7 (BRD7) pathway for human osteosarcoma. Oncotarget. 5:3088–3100. 2014. View Article : Google Scholar : | |
Liu C, Xiong Q, Li Q, Lin W, Jiang S, Zhang D, Wang Y, Duan X, Gong P and Kang N: CHD7 regulates bone-fat balance by suppressing PPAR-γ signaling. Nat Commun. 13:19892022. View Article : Google Scholar | |
Takada I, Yogiashi Y and Kato S: Signaling crosstalk between PPARγ and BMP2 in mesenchymal stem cells. PPAR Res. 2012:6071412012. View Article : Google Scholar | |
Schnetz MP, Bartels CF, Shastri K, Balasubramanian D, Zentner GE, Balaji R, Zhang X, Song L, Wang Z, Laframboise T, et al: Genomic distribution of CHD7 on chromatin tracks H3K4 methylation patterns. Genome Res. 19:590–601. 2009. View Article : Google Scholar : PubMed/NCBI | |
Newton AH and Pask AJ: CHD9 upregulates RUNX2 and has a potential role in skeletal evolution. BMC Mol Cell Biol. 21:272020. View Article : Google Scholar : PubMed/NCBI | |
Zhou C, Zou J, Zou S and Li X: INO80 is required for osteogenic differentiation of human mesenchymal stem cells. Sci Rep. 6:359242016. View Article : Google Scholar : PubMed/NCBI | |
Anwar A, Sapra L, Gupta N, Ojha RP, Verma B and Srivastava RK: Fine-tuning osteoclastogenesis: An insight into the cellular and molecular regulation of osteoclastogenesis. J Cell Physiol. 238:1431–1464. 2023. View Article : Google Scholar : PubMed/NCBI | |
Du J, Liu Y, Sun J, Yao E, Xu J, Wu X, Xu L, Zhou M, Yang G and Jiang X: ARID1A safeguards the canalization of the cell fate decision during osteoclastogenesis. Nat Commun. 15:59942024. View Article : Google Scholar : PubMed/NCBI | |
Urban W, Krzystańska D, Piekarz M, Nazar J and Jankowska A: Osteosarcoma's genetic landscape painted by genes' mutations. Acta Biochim Pol. 70:671–678. 2023.PubMed/NCBI | |
Gaeta R, Morelli M, Lessi F, Mazzanti CM, Menicagli M, Capanna R, Andreani L, Coccoli L, Aretini P and Franchi A: Identification of new potential prognostic and predictive markers in high-grade osteosarcoma using whole exome sequencing. Int J Mol Sci. 24:100862023. View Article : Google Scholar : PubMed/NCBI | |
Tuckermann J and Adams RH: The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat Rev Rheumatol. 17:608–620. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bixel MG, Sivaraj KK, Timmen M, Mohanakrishnan V, Aravamudhan A, Adams S, Koh BI, Jeong HW, Kruse K, Stange R and Adams RH: Angiogenesis is uncoupled from osteogenesis during calvarial bone regeneration. Nat Commun. 15:45752024. View Article : Google Scholar : PubMed/NCBI | |
Duan X, Murata Y, Liu Y, Nicolae C, Olsen BR and Berendsen AD: Vegfa regulates perichondrial vascularity and osteoblast differentiation in bone development. Development. 142:1984–1991. 2015. View Article : Google Scholar : PubMed/NCBI | |
Sena JA, Wang L and Hu CJ: BRG1 and BRM chromatin-remodeling complexes regulate the hypoxia response by acting as coactivators for a subset of hypoxia-inducible transcription factor target genes. Mol Cell Biol. 33:3849–3863. 2013. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Zhang R, Beischlag TV, Muchardt C, Yaniv M and Hankinson O: Roles of Brahma and Brahma/SWI2-related gene 1 in hypoxic induction of the erythropoietin gene. J Biol Chem. 279:46733–46741. 2004. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Chen Y, Bao L, Zhang B, Wang JE, Kumar A, Xing C, Wang Y and Luo W: CHD4 promotes breast cancer progression as a coactivator of hypoxia-inducible factors. Cancer Res. 80:3880–3891. 2020. View Article : Google Scholar : PubMed/NCBI | |
Collins JM, Lang A, Parisi C, Moharrer Y, Nijsure MP, Thomas Kim JH, Ahmed S, Szeto GL, Qin L, Gottardi R, et al: YAP and TAZ couple osteoblast precursor mobilization to angiogenesis and mechanoregulation in murine bone development. Dev Cell. 59:211–227.e5. 2024. View Article : Google Scholar : | |
Griffin CT, Brennan J and Magnuson T: The chromatin-remodeling enzyme BRG1 plays an essential role in primitive erythropoiesis and vascular development. Development. 135:493–500. 2008. View Article : Google Scholar | |
Davis RB, Curtis CD and Griffin CT: BRG1 promotes COUP-TFII expression and venous specification during embryonic vascular development. Development. 140:1272–1281. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ingram KG, Curtis CD, Silasi-Mansat R, Lupu F and Griffin CT: The NuRD chromatin-remodeling enzyme CHD4 promotes embryonic vascular integrity by transcriptionally regulating extracellular matrix proteolysis. PLoS Genet. 9:e10040312013. View Article : Google Scholar : PubMed/NCBI | |
Forriol F and Shapiro F: Bone development: interaction of molecular components and biophysical forces. Clin Orthop Relat Res. 432:14–33. 2005. View Article : Google Scholar | |
Komori T: Cell death in chondrocytes, osteoblasts, and osteocytes. Int J Mol Sci. 17:20452016. View Article : Google Scholar : PubMed/NCBI | |
Grandy R, Sepulveda H, Aguilar R, Pihan P, Henriquez B, Olate J and Montecino M: The Ric-8B gene is highly expressed in proliferating preosteoblastic cells and downregulated during osteoblast differentiation in a SWI/SNF- and C/EBPbeta-mediated manner. Mol Cell Biol. 31:2997–3008. 2011. View Article : Google Scholar : PubMed/NCBI | |
Zhang M, Guo T, Pei F, Feng J, Jing J, Xu J, Yamada T, Ho TV, Du J, Sehgal P and Chai Y: ARID1B maintains mesenchymal stem cell quiescence via inhibition of BCL11B-mediated non-canonical activin signaling. Nat Commun. 15:46142024. View Article : Google Scholar : PubMed/NCBI | |
Ho L, Ronan JL, Wu J, Staahl BT, Chen L, Kuo A, Lessard J, Nesvizhskii AI, Ranish J and Crabtree GR: An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency. Proc Natl Acad Sci USA. 106:5181–5186. 2009. View Article : Google Scholar : PubMed/NCBI | |
Schaniel C, Ang YS, Ratnakumar K, Cormier C, James T, Bernstein E, Lemischka IR and Paddison PJ: Smarcc1/Baf155 couples self-renewal gene repression with changes in chromatin structure in mouse embryonic stem cells. Stem Cells. 27:2979–2991. 2009. View Article : Google Scholar : PubMed/NCBI | |
Yan Z, Wang Z, Sharova L, Sharov AA, Ling C, Piao Y, Aiba K, Matoba R, Wang W and Ko MS: BAF250B-associated SWI/SNF chromatin-remodeling complex is required to maintain undifferentiated mouse embryonic stem cells. Stem Cells. 26:1155–1165. 2008. View Article : Google Scholar : PubMed/NCBI | |
Eleuteri B, Aranda S and Ernfors P: NoRC Recruitment by H2A.X deposition at rRNA gene promoter limits embryonic stem cell proliferation. Cell Rep. 23:1853–1866. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li B, Lyu P, Tang J, Li J, Ouchi T, Fan Y, Zhao Z and Li L: The potential role and therapeutic relevance of cellular senescence in skeletal pathophysiology. J Gerontol A Biol Sci Med Sci. 79:glae0372024. View Article : Google Scholar : PubMed/NCBI | |
Napolitano MA, Cipollaro M, Cascino A, Melone MA, Giordano A and Galderisi U: Brg1 chromatin remodeling factor is involved in cell growth arrest, apoptosis and senescence of rat mesenchymal stem cells. J Cell Sci. 120(Pt 16): 2904–2911. 2007. View Article : Google Scholar : PubMed/NCBI | |
Alessio N, Squillaro T, Cipollaro M, Bagella L, Giordano A and Galderisi U: The BRG1 ATPase of chromatin remodeling complexes is involved in modulation of mesenchymal stem cell senescence through RB-P53 pathways. Oncogene. 29:5452–5463. 2010. View Article : Google Scholar : PubMed/NCBI | |
Squillaro T, Severino V, Alessio N, Farina A, Di Bernardo G, Cipollaro M, Peluso G, Chambery A and Galderisi U: De-regulated expression of the BRG1 chromatin remodeling factor in bone marrow mesenchymal stromal cells induces senescence associated with the silencing of NANOG and changes in the levels of chromatin proteins. Cell Cycle. 14:1315–1326. 2015. View Article : Google Scholar : PubMed/NCBI | |
Yuan J and Ofengeim D: A guide to cell death pathways. Nat Rev Mol Cell Biol. 25:379–395. 2024. View Article : Google Scholar | |
Klochendler-Yeivin A, Fiette L, Barra J, Muchardt C, Babinet C and Yaniv M: The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression. EMBO Rep. 1:500–506. 2000. View Article : Google Scholar | |
Li S, Huang Z, Zhu Y, Yan J, Li J, Chen J, Zhou J, Zhang Y, Chen W, Xu K and Ye W: Bromodomain-containing protein 7 regulates matrix metabolism and apoptosis in human nucleus pulposus cells through the BRD7-PI3K-YAP1 signaling axis. Exp Cell Res. 405:1126582021. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z and Li N: Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res. 71:227–262. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chai S, Yang Y, Wei L, Cao Y, Ma J, Zheng X, Teng J and Qin N: Luteolin rescues postmenopausal osteoporosis elicited by OVX through alleviating osteoblast pyroptosis via activating PI3K-AKT signaling. Phytomedicine. 128:1555162024. View Article : Google Scholar : PubMed/NCBI | |
Ruan H, Zhang H, Feng J, Luo H, Fu F, Yao S, Zhou C, Zhang Z, Bian Y, Jin H, et al: Inhibition of Caspase-1-mediated pyroptosis promotes osteogenic differentiation, offering a therapeutic target for osteoporosis. Int Immunopharmacol. 124(Pt B): 1109012023. View Article : Google Scholar : PubMed/NCBI | |
Deng Z, Zhang Y, Zhu Y, Zhu J, Li S, Huang Z, Qin T, Wu J, Zhang C, Chen W, et al: BRD9 inhibition attenuates matrix degradation and pyroptosis in nucleus pulposus by modulating the NOX1/ROS/NF-κB axis. Inflammation. 46:1002–1021. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Zhang Y, Cao J, Wang Y, Anwar N, Zhang Z, Zhang D, Ma Y, Xiao Y, Xiao L and Wang X: The role of autophagy in bone metabolism and clinical significance. Autophagy. 19:2409–2427. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ren X, Xu J, Xue Q, Tong Y, Xu T, Wang J, Yang T, Chen Y, Shi D and Li X: BRG1 enhances porcine iPSC pluripotency through WNT/β-catenin and autophagy pathways. Theriogenology. 215:10–23. 2024. View Article : Google Scholar | |
Yang C, Tian Y, Zhao F, Chen Z, Su P, Li Y and Qian A: Bone microenvironment and osteosarcoma metastasis. Int J Mol Sci. 21:69852020. View Article : Google Scholar : PubMed/NCBI | |
Urlić I, Jovičić MŠ, Ostojić K and Ivković A: Cellular and genetic background of osteosarcoma. Curr Issues Mol Biol. 45:4344–4358. 2023. View Article : Google Scholar | |
Hang JF and Chen PC: Parosteal osteosarcoma. Arch Pathol Lab Med. 138:694–699. 2014. View Article : Google Scholar : PubMed/NCBI | |
Qiu YQ and Chen YL: Primary meningeal osteoblastic osteosarcoma containing fibroblast osteosarcoma: Clinicopathological analysis and literature review. Osteoporos Int. 32:1007–1012. 2021. View Article : Google Scholar | |
Guan Y, Zhang W, Mao Y and Li S: Nanoparticles and bone microenvironment: A comprehensive review for malignant bone tumor diagnosis and treatment. Mol Cancer. 23:2462024. View Article : Google Scholar : PubMed/NCBI | |
Basu Mallick A and Chawla SP: Giant cell tumor of bone: An update. Curr Oncol Rep. 23:512021. View Article : Google Scholar : PubMed/NCBI | |
Corre I, Verrecchia F, Crenn V, Redini F and Trichet V: The osteosarcoma microenvironment: A complex but targetable ecosystem. Cells. 9:9762020. View Article : Google Scholar : PubMed/NCBI | |
Grünewald TGP, Cidre-Aranaz F, Surdez D, Tomazou EM, de Álava E, Kovar H, Sorensen PH, Delattre O and Dirksen U: Ewing sarcoma. Nat Rev Dis Primers. 4:52018. View Article : Google Scholar : PubMed/NCBI | |
Weber K, Damron TA, Frassica FJ and Sim FH: Malignant bone tumors. Instr Course Lect. 57:673–688. 2008.PubMed/NCBI | |
Graca Marques J, Pavlovic B, Ngo QA, Pedot G, Roemmele M, Volken L, Kisele S, Perbet R, Wachtel M and Schäfer BW: The chromatin remodeler CHD4 sustains ewing sarcoma cell survival by controlling global chromatin architecture. Cancer Res. 84:241–257. 2024. View Article : Google Scholar | |
Sohn EJ and Libich DS: Hijacking the BAF complex: The mechanistic interplay of ARID1A and EWS::FLI1 in Ewing sarcoma. Mol Oncol. 19:961–964. 2025. View Article : Google Scholar : | |
Selvanathan SP, Graham GT, Grego AR, Baker TM, Hogg JR, Simpson M, Batish M, Crompton B, Stegmaier K, Tomazou EM, et al: EWS-FLI1 modulated alternative splicing of ARID1A reveals novel oncogenic function through the BAF complex. Nucleic Acids Res. 47:9619–9636. 2019.PubMed/NCBI | |
Boulay G, Sandoval GJ, Riggi N, Iyer S, Buisson R, Naigles B, Awad ME, Rengarajan S, Volorio A, McBride MJ, et al: Cancer-specific retargeting of BAF complexes by a prion-like domain. Cell. 171:163–178.e19. 2017. View Article : Google Scholar : PubMed/NCBI | |
Jayabal P, Zhou F, Lei X, Ma X, Blackman B, Weintraub ST, Houghton PJ and Shiio Y: NELL2-cdc42 signaling regulates BAF complexes and Ewing sarcoma cell growth. Cell Rep. 36:1092542021. View Article : Google Scholar : PubMed/NCBI | |
Cyra M, Schulte M, Berthold R, Heinst L, Jansen EP, Grünewald I, Elges S, Larsson O, Schliemann C, Steinestel K, et al: SS18-SSX drives CREB activation in synovial sarcoma. Cell Oncol (Dordr). 45:399–413. 2022. View Article : Google Scholar : PubMed/NCBI | |
Michel BC, D'Avino AR, Cassel SH, Mashtalir N, McKenzie ZM, McBride MJ, Valencia AM, Zhou Q, Bocker M, Soares LMM, et al: A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nat Cell Biol. 20:1410–1420. 2018. View Article : Google Scholar : PubMed/NCBI | |
Shih AR, Cote GM, Chebib I, Choy E, DeLaney T, Deshpande V, Hornicek FJ, Miao R, Schwab JH, Nielsen GP and Chen YL: Clinicopathologic characteristics of poorly differentiated chordoma. Mod Pathol. 31:1237–1245. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sergi CM: Commentary on: SMARCB1 as a novel diagnostic and prognostic biomarker for osteosarcoma. Biosci Rep. 42:BSR202200402022. View Article : Google Scholar : PubMed/NCBI | |
Mobley BC, McKenney JK, Bangs CD, Callahan K, Yeom KW, Schneppenheim R, Hayden MG, Cherry AM, Gokden M, Edwards MS, et al: Loss of SMARCB1/INI1 expression in poorly differentiated chordomas. Acta Neuropathol. 120:745–753. 2010. View Article : Google Scholar : PubMed/NCBI | |
Ding J, Yu C, Sui Y, Wang L, Yang Y, Wang F, Yao H, Xing F, Liu H, Li Y, et al: The chromatin remodeling protein INO80 contributes to the removal of H2A.Z at the p53-binding site of the p21 gene in response to doxorubicin. FEBS J. 285:3270–3285. 2018. View Article : Google Scholar : PubMed/NCBI | |
Flowers S, Beck GR Jr and Moran E: Transcriptional activation by pRB and its coordination with SWI/SNF recruitment. Cancer Res. 70:8282–8287. 2010. View Article : Google Scholar : PubMed/NCBI | |
Meisenberg C, Pinder SI, Hopkins SR, Wooller SK, Benstead-Hume G, Pearl FMG, Jeggo PA and Downs JA: Repression of transcription at DNA breaks requires cohesin throughout interphase and prevents genome instability. Mol Cell. 73:212–223.e7. 2019. View Article : Google Scholar : | |
Ivanov AV, Peng H, Yurchenko V, Yap KL, Negorev DG, Schultz DC, Psulkowski E, Fredericks WJ, White DE, Maul GG, et al: PHD domain-mediated E3 ligase activity directs intramolecular sumoylation of an adjacent bromodomain required for gene silencing. Mol Cell. 28:823–837. 2007. View Article : Google Scholar : PubMed/NCBI | |
Walhart TA, Vacca B, Hepperla AJ, Hamad SH, Petrongelli J, Wang Y, McKean EL, Moksa M, Cao Q, Yip S, et al: SMARCB1 loss in poorly differentiated chordomas drives tumor progression. Am J Pathol. 193:456–473. 2023. View Article : Google Scholar : PubMed/NCBI | |
Passeri T, Gutman T, Hamza A, Adle-Biassette H, Girard E, Beaurepere R, Tariq Z, Mariani O, Dahmani A, Bourneix C, et al: The mutational landscape of skull base and spinal chordomas and the identification of potential prognostic and theranostic biomarkers. J Neurosurg. 139:1270–1280. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wu X, Lin X, Chen Y, Kong W, Xu J and Yu Z: Response of metastatic chordoma to the immune checkpoint inhibitor pembrolizumab: A case report. Front Oncol. 10:5659452020. View Article : Google Scholar | |
Wang L, Zehir A, Nafa K, Zhou N, Berger MF, Casanova J, Sadowska J, Lu C, Allis CD, Gounder M, et al: Genomic aberrations frequently alter chromatin regulatory genes in chordoma. Genes Chromosomes Cancer. 55:591–600. 2016. View Article : Google Scholar : PubMed/NCBI | |
Harlow ML, Chasse MH, Boguslawski EA, Sorensen KM, Gedminas JM, Kitchen-Goosen SM, Rothbart SB, Taslim C, Lessnick SL, Peck AS, et al: Trabectedin inhibits EWS-FLI1 and evicts SWI/SNF from chromatin in a schedule-dependent manner. Clin Cancer Res. 25:3417–3429. 2019. View Article : Google Scholar : PubMed/NCBI | |
Livingston JA, Blay JY, Trent J, Valverde C, Agulnik M, Gounder M, Le Cesne A, McKean M, Wagner MJ, Stacchiotti S, et al: A phase I study of FHD-609, a heterobifunctional degrader of bromodomain-containing protein 9, in patients with advanced synovial sarcoma or SMARCB1-deficient tumors. Clin Cancer Res. 31:628–638. 2025. View Article : Google Scholar | |
Dreier MR, Walia J and de la Serna IL: Targeting SWI/SNF complexes in cancer: Pharmacological approaches and implications. Epigenomes. 8:72024. View Article : Google Scholar : PubMed/NCBI | |
Gatchalian J, Liao J, Maxwell MB and Hargreaves DC: Control of stimulus-dependent responses in macrophages by SWI/SNF chromatin remodeling complexes. Trends Immunol. 41:126–140. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li X, Ding D, Yao J, Zhou B, Shen T, Qi Y, Ni T and Wei G: Chromatin remodeling factor BAZ1A regulates cellular senescence in both cancer and normal cells. Life Sci. 229:225–232. 2019. View Article : Google Scholar : PubMed/NCBI | |
Martel-Pelletier J, Barr AJ, Cicuttini FM, Conaghan PG, Cooper C, Goldring MB, Goldring SR, Jones G, Teichtahl AJ, Pelletier JP, et al: Osteoarthritis. Nat Rev Dis Primers. 2:160722016. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Zhang Y, Zhang L, He D, Zhao L, Miao Z, Cheng W, Zhu C, Zhu L, Zhang W, et al: IRF1 governs the expression of SMARCC1 via the GCN5-SETD2 axis and actively engages in the advancement of osteoarthritis. J Orthop Translat. 45:211–225. 2024. View Article : Google Scholar : PubMed/NCBI | |
Qiu Y, Yao J, Li L, Xiao M, Meng J, Huang X, Cai Y, Wen Z, Huang J, Zhu M, et al: Machine learning identifies ferroptosis-related genes as potential diagnostic biomarkers for osteoarthritis. Front Endocrinol (Lausanne). 14:11987632023. View Article : Google Scholar : PubMed/NCBI | |
Devlin MJ and Rosen CJ: The bone-fat interface: Basic and clinical implications of marrow adiposity. Lancet Diabetes Endocrinol. 3:141–147. 2015. View Article : Google Scholar | |
Kuznia AL, Hernandez AK and Lee LU: Adolescent idiopathic scoliosis: Common questions and answers. Am Fam Physician. 101:19–23. 2020.PubMed/NCBI | |
Cheng JC, Castelein RM, Chu WC, Danielsson AJ, Dobbs MB, Grivas TB, Gurnett CA, Luk KD, Moreau A, Newton PO, et al: Adolescent idiopathic scoliosis. Nat Rev Dis Primers. 1:150302015. View Article : Google Scholar : PubMed/NCBI | |
Wu Z, Dai Z, Yuwen W, Liu Z, Qiu Y, Cheng JC, Zhu Z and Xu L: Genetic variants of CHD7 are associated with adolescent idiopathic scoliosis. Spine (Phila Pa 1976). 46:E618–E624. 2021. View Article : Google Scholar | |
Borysiak K, Janusz P, Andrusiewicz M, Chmielewska M, Kozinoga M, Kotwicki T and Kotwicka M: CHD7 gene polymorphisms in female patients with idiopathic scoliosis. BMC Musculoskelet Disord. 21:182020. View Article : Google Scholar : PubMed/NCBI | |
Kulkarni S, Nagarajan P, Wall J, Donovan DJ, Donell RL, Ligon AH, Venkatachalam S and Quade BJ: Disruption of chromodomain helicase DNA binding protein 2 (CHD2) causes scoliosis. Am J Med Genet A. 146A:1117–1127. 2008. View Article : Google Scholar : PubMed/NCBI | |
Bonyadi M, Waldman SD, Liu D, Aubin JE, Grynpas MD and Stanford WL: Mesenchymal progenitor self-renewal deficiency leads to age-dependent osteoporosis in Sca-1/Ly-6A null mice. Proc Natl Acad Sci USA. 100:5840–5845. 2003. View Article : Google Scholar : PubMed/NCBI | |
Yu W, Wang HL, Zhang J and Yin C: The effects of epigenetic modifications on bone remodeling in age-related osteoporosis. Connect Tissue Res. 64:105–116. 2023. View Article : Google Scholar | |
Li C, Pan H, Liu W, Jin G, Liu W, Liang C and Jiang X: Discovery of novel serum biomarkers for diagnosing and predicting postmenopausal osteoporosis patients by 4D-label free protein omics. J Orthop Res. 41:2713–2720. 2023. View Article : Google Scholar : PubMed/NCBI | |
Pico MJ, Hashemi S, Xu F, Nguyen KH, Donnelly R, Moran E and Flowers S: Glucocorticoid receptor-mediated cis-repression of osteogenic genes requires BRM-SWI/SNF. Bone Rep. 5:222–227. 2016. View Article : Google Scholar | |
Chi SN, Yi JS, Williams PM, Roy-Chowdhuri S, Patton DR, Coffey BD, Reid JM, Piao J, Saguilig L, Alonzo TA, et al: Tazemetostat for tumors harboring SMARCB1/SMARCA4 or EZH2 alterations: Results from NCI-COG pediatric MATCH APEC1621C. J Natl Cancer Inst. 115:1355–1363. 2023. View Article : Google Scholar : PubMed/NCBI | |
Martin LJ, Koegl M, Bader G, Cockcroft XL, Fedorov O, Fiegen D, Gerstberger T, Hofmann MH, Hohmann AF, Kessler D, et al: Structure-based design of an in vivo active selective BRD9 inhibitor. J Med Chem. 59:4462–4475. 2016. View Article : Google Scholar : PubMed/NCBI |