Open Access

Role of ubiquitination in the occurrence and development of osteoporosis (Review)

  • Authors:
    • Xiaoxia Fan
    • Rong Zhang
    • Guocai Xu
    • Peiyun Fan
    • Wei Luo
    • Chunmei Cai
    • Ri-Li Ge
  • View Affiliations

  • Published online on: June 26, 2024     https://doi.org/10.3892/ijmm.2024.5392
  • Article Number: 68
  • Copyright : © Fan et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The ubiquitin (Ub)‑proteasome system (UPS) plays a pivotal role in maintaining protein homeostasis and function to modulate various cellular processes including skeletal cell differentiation and bone homeostasis. The Ub ligase E3 promotes the transfer of Ub to the target protein, especially transcription factors, to regulate the proliferation, differentiation and survival of bone cells, as well as bone formation. In turn, the deubiquitinating enzyme removes Ub from modified substrate proteins to orchestrate bone remodeling. As a result of abnormal regulation of ubiquitination, bone cell differentiation exhibits disorder and then bone homeostasis is affected, consequently leading to osteoporosis. The present review discussed the role and mechanism of UPS in bone remodeling. However, the specific mechanism of UPS in the process of bone remodeling is still not fully understood and further research is required. The study of the mechanism of action of UPS can provide new ideas and methods for the prevention and treatment of osteoporosis. In addition, the most commonly used osteoporosis drugs that target ubiquitination processes in the clinic are discussed in the current review.

1. Introduction

There are >8.9 million osteoporotic fractures occurring annually in the world (1). A total of >33% of women and 20% of men >50 years old will suffer an osteoporotic fracture at least once in their lifetime (2). With the increase in population aging, osteoporosis has become a chronic disease involving fractures, high disability and mortality and increased social and economic burden (3). Osteoporosis is a multifactorial disease characterized by bone loss and damage to the microstructure of the trabecular bone, leading to an increased risk of fracture and bone fragility (4,5). Therefore, identifying the pathogenesis of osteoporosis is of high importance.

Osteoporosis is closely linked to imbalances in bone homeostasis. The maintenance of bone homeostasis mainly depends on the balance between osteoclasts and osteoblasts; osteoclasts mediate bone resorption and osteoblasts mediate bone formation. In organisms, bone tissue constantly undergoes remodeling process through the synergistic action of bone marrow mesenchymal cells (BMSCs), osteoblasts, osteocytes and osteoclasts (6). BMSCs can multi-directionally differentiate into osteogenesis, adipogenesis and cartilage. BMSCs also promote calcium deposition and bone formation (7). Osteoblasts are derived from BMSCs and play a key role in the process of bone formation. They are mainly distributed on the surface of bone and synthesize bone matrix by secreting collagen and bone matrix proteins. Osteoblasts can synthesize and secrete the main components of bone, including collagen fibers, chondroitin sulfate, phosphoric acid and calcium. These secretions are important components of the bone matrix and play an important role in maintaining the structure and function of bone. Osteoclasts, originating from the hematopoietic monocyte-macrophage system, are a special type of terminally differentiated cells. They are involved in the process of bone remodeling through the uptake of bone matrix and minerals, as well as the secretion of organic acids and protein hydrolyzing enzymes, thus maintaining the normal structure and function of the skeleton (8). As osteoblast-derived cells, osteocytes maintain mature bone metabolism by mediating cytokine sensing, transduction and secretion to modulate bone resorption and formation (9). Under pathological conditions, osteoclast activity increases, while osteogenesis is either weakened or impaired, consequently destroying bone homeostasis. Over time, osteoporosis develops further.

During bone homeostasis and repair, Wnt/β-catenin, Hedgehog, bone morphogenetic protein (BMP-2)/Smad and phosphatidylinositol-3-kinase/protein kinase B and others are the major regulatory signaling pathways during skeletal growth and development. The Wnt/β-catenin signaling pathway plays a key regulatory role in bone development and formation by affecting the proliferation and differentiation of osteoblasts and osteoclast generation. Wnt is composed of 19 secreted glycoproteins and has the function of regulating cell growth, differentiation and apoptosis (10,11). The receptor activator of nuclear factor-κB ligand (RANK-L)/RANK/MAPK and the NF-κB axes are the major regulatory signaling pathways in osteoclast formation and function. RANKL integrates with the osteoclast surface receptor RANK, which in turn recruits tumor necrosis factor receptor-associated factor (TRAF) 6 and further activates multiple downstream signaling pathways, such as three mitogen-activated protein kinases (MAPKs) including p38 kinase (p38MAPK), extracellular signal-regulated kinase (ERK) and C-Jun N-terminal kinase (12,13).

Ubiquitination is a conserved, widespread and dynamic post-translational modification involved in regulating the activity and stability of thousands of proteins, thereby affecting most cellular functions and responses (14). Ubiquitylation is a process mediated by the covalent attachment of ubiquitin (Ub) to the target protein, catalyzed by a series of enzymatic cascades, including E1-mediating activation, E2-mediating coupling and E3-mediating connection step (Fig. 1) (15). E3 then targets the protein by promoting the formation of an isopeptide bond between the C-terminal carboxyl group of Ub and one of the seven lysine residues of Ub (K6/11/27/29/33/48/63) or the N-terminal methionine of Ub (M1) (16-18). Subsequently, modified substrate proteins are degraded by 26S protein complexes or exhibit cellular function (19,20). E3 provides the reaction rate and substrate specificity for this cascade reaction. Accumulating evidence has shown that E3 ligases are associated with a number of biological processes, including bone homeostasis (21). There are 600-1,000 E3 ligases encoded by the human genome. According to the domain structure and ubiquitination mechanism through which E3 ligases interact with target proteins, E3 members are broadly divided into three major subfamilies: i) Homology to E6AP C-Terminus (HECT) family; ii) really interesting new gene figure (RING) family; and iii) RING-between-RING (RBR; Fig. 2). The reversibility of ubiquitination modification is achieved by deubiquitinating enzymes (DUBs) (22). At present, >100 DUBs are encoded in the human genome based on the catalytic region. They can be classified into two categories or six subfamilies (Fig. 3) (23-28).

Figure 2

Classification of the Ub ligase E3. The HECT ligase can be divided into three subgroups based on the existence of the different domains it contains: i) NEDD4 family, including the C2 domain, 2-4 WW domain and HECT; ii) HERC family, including HECT and RCC1-like domains, E3 at least one or more RLDs; and iii) other families containing variable number and types of domains. All members of the RNF family are characterized by the N-terminal RING domain. Each subfamily has its unique domains besides the conserved RING domain. The MARCH subfamily is characterized by the zero to more C-terminal TM domain. The PA-TM-RING subfamily is characterized by the PA domain, two TM domains and longer C-end tail extension. The RBR subfamily is characterized by the IBR domain. The TRIM subfamily is characterized by two BB domains and a CC domain. The UIM subfamily is characterized by a C2HC-type zinc finger, two C2H2-type zinc fingers and UIM. The name RBR derives from the existence of two RING domains, RING1 and RING2, which are separated by the IBR. Each member of the RBR E3 ligase contains other specific domains, such as Parkin, which also contains RING0 and Ub domains. Ub, ubiquitin; HECT, homology to E6AP C-Terminus; NEDD4, neural precursor cell expressed developmentally downregulated protein 4; RLD, RCC1-like domain; RING, really interesting new gene figure; MARCH, Membrane-associated RING-CH; PA, protease-associated; TM, transmembrane; RBR, RING-between-RING; IBR, in-between-RING domain; UIM, ubiquitin-interacting motifs.

In recent years, a growing body of evidence suggests that ubiquitination plays an important regulatory role in the maintenance of bone homeostasis. Osteoporosis and other related bone diseases are accompanied by abnormal expression and dysfunction of ubiquitination and de-ubiquitination enzymes. As a potential research hotspot for bone metabolism-related diseases and a new target for drug development, ubiquitination has a broad prospect in the field of osteoporosis prevention and treatment. The current review discusses the specific mechanism and role of ubiquitination in bone remodeling to provide a more theoretical basis and practical guidance for the treatment of related diseases (Fig. 4; Table I).

Table I

E3 ubiquitin ligases, deubiquitinases and bone remodeling.

Table I

E3 ubiquitin ligases, deubiquitinases and bone remodeling.

FamilyEnzymeMechanismFunction(Refs.)
HECT familySmurf1Interacts with Runx2 in a ubiquitin proteasome dependent manner, mediating its degradationInhibits osteoblast differentiation and mineralization(27)
Degrades BMP downstream signaling molecules(28)
Control JunB turnover(29)
Involved in P53/miR-17/Smurf1 pathway(30)
Smurf2Ubiquitinates TGF-β receptor and Smad proteinsInhibits osteoblast differentiation and proliferation and enhances osteoclast formation(33,34)
Inhibits the BMP/Smad signaling pathwayInhibits osteogenesis(35)
Information transmission between osteoblasts and osteoclastRegulates the activity of osteoblast dependent osteoclast(32)
WWP1Mediates Runx2 degradation(36,37)
Targets JunB for ubiquitination and degradation in MSCsInhibits osteogenic differentiation(38)
WWP2Mediates mono-ubiquitination of Runx2Enhances osteogenic differentiation(39)
ITCHMechanism unknownInhibits osteogenic differentiation(45,46)
Limits TRAF6 deubiquitination by binding the cylindromatosisInhibits osteoclastogenesis(47)
NEDD4Induces degradation of TGF-1β-activated pSMAD1 and improves the activation of pSMAD2 and pERK1/2 pathwaysEnhances osteoblast proliferation and bone mass accumulation(54-56)
RING familyRNF146Mediates the PARsylation of Tankyrase to stimulate Wnt pathway via degrading Axin.Enhances osteogenic differentiation(40,41)
Regulates the stability of SRC and 3BP2, the adaptor protein essential for OCsInhibits osteoclastogenesis(42)
RNF185Regulates Wnt signaling pathway by degrading dishevelled 2Inhibit osteogenic differentiation(43,44)
SKP2Negatively targets Runx2 for ubiquitin-mediated degradationInhibits osteogenic differentiation(48,49)
MURF1Mechanism unknownPromotes osteoclastogenesis(50,51)
TRAF4Degrades Smurf2Promotes osteogenic differentiation(53)
Ubiquitin conjugating enzyme USPUBE2E3Might regulate the nuclear factor erythroid 2-related factor and control its functionPromotes osteogenic differentiation(52)
USP1Acts on TRAF6 by suppressing TRAF6 ubiquitination to control NF-κB signalingInhibits osteoblastic pyroptosis(61)
Deubiquitinated ID proteins promote the preservation of mesenchymal stem cell signaturesInhibits osteogenic differentiation(62)
USP4Through Dishevelled inhibits
Wnt/β-catenin signal transduction
Antagonizes osteoblast differentiation(67,68)
Targets TGF β receptor to enhance the activation of TGF/BMP signaling pathwayPromotes the proliferation and differentiation of MSCs(69,70)
USP7Mechanism unknownPromotes osteogenic differentiation of hASCs(60)
Binds to and deubiquitination of HMGB1Promotes osteoclast differentiation(87)
USP8Attenuates TRAF6/TAK1 Axis and stimulates STING SignalingInhibits osteoclastogenesis(88)
Prevents the degradation of the Frizzy-5 ubiquitinated Wnt receptor to ensure
Wnt-induced osteogenesis
Promotes osteogenesis(63)
USP11Stabilizes the MSX1 proteinPromotes the osteogenic differentiation(64)
USP18Regulates Type I IFN signaling by deconjugating ISGlationInhibits osteoclastogenesis(89-91)
USP26Stabilizes β-catenin to promote the osteogenic activity of MSCsPromotes the osteogenic differentiation(93)
Enhances IκBα stability to inhibit NF-κB activationSuppress osteoclast differentiation
USP34Modulates BMP2 signaling in mesenchymal stem cellsPromotes osteogenic differentiation and bone formation(66)
Inhibits the NF-κB pathway by deubiquitinating and stabling IκBαInhibits osteoclastogenesis(92)
USP53Interacts with F-box protein 31 to promote the degradation of β-catenin by Skp1/Cul1/F-box protein complexEnhances the osteogenic and differentiation(71)
Act on vitamin D-receptor-SMAD3 pathwayPromotes osteogenesis(72)
CYLDNegatively regulates the RANK signaling pathway and osteoclast formation by deubiquitinating and inactivating TRAF6Inhibits osteoclastogenesis(75-77)
UCHsUCHL1Regulates BMP2/Smad signal pathwayPromotes osteogenesis(73,74)
UCHL3Regulates BMP2 induced Smad1 polyubiquitinationPromotes osteoblast differentiation(66)
OTUsA20Attenuates RANKL inducing NF-κB signaling by altering IκB ubiquitination-mediated degradationInhibits osteoclast differentiation(78-81)
Via TRAF6-dependent autophagy in under hypoxiaInhibits osteoclastogenesis(82)
OTUB1Stabilizes FGFR2Promotes osteogenesis(94)
JAMM/MPNMYSM1Mechanism unknownPromotes osteogenic differentiation(85)
Mechanism unknownIncreases osteoclast number and absorption activity(86)

[i] HECT, homology to E6AP C-Terminus; Runx, Runt-related transcription factor 2; BMP, bone morphogenetic protein; JunB, Jun B proto-oncogene; miR, microRNA; Smurf, Smad ubiquitination regulatory factor; WWP, WW domain-containing E3 Ub protein ligase; MSCs, mesenchymal cells; ITCH, itchy E3 ubiquitin protein ligase; TRAF, tumor necrosis factor receptor-associated factor; NEDD4, neural precursor cell expressed developmentally downregulated protein 4; RING, really interesting new gene figure; RNF, The RING finger; OCs, osteoclasts, SKP, S-phase kinase-associated protein; MURF, Muscle-specific RING finger E3 ligase; UBE2E3, Ubiquitin-conjugating enzyme E2 E3; USP, Ub-specific proteases; HMGB1, high mobility group box 1; TAK1, Transforming growth factor-β (TGF-β)-activated kinase 1; STING, stimulator of interferon genes; MSX1, Msh Homeobox 1; RANK, receptor activator of nuclear factor-κB; RANKL, RANK ligand; ovarian tumor domain-containing ubiquitin aldehyde-binding protein 1; FGFR2, Fibroblast Growth Factor Receptor; JAMM, Jab1/MPN domain associated metalloisopeptidase; MPN, Mpr1/Pad1 N-terminal; MYSM1, MPN domain containing 1.

2. E3 Ub ligases and the bone

In a growing number of studies, bone remodeling has been linked to ubiquitination. E3 Ub ligases are mainly involved in osteoblast differentiation and bone formation. In the present review, the function and regulation of E3 ligases in bone homeostasis is discussed.

Smad ubiquitination regulatory factor (Smurf) 1

Belonging to the C2-WW-HECT region Ub ligases, Smurf1 downregulates the level of alkaline phosphatase and osteocalcin and suppresses the osteogenic activity of osteoblasts, resulting in osteoporosis and fragility fractures. Al-Rawi et al (29) found that the micro duplication of the Smurf1 gene led to the first case of childhood osteoporosis. Mechanically, Smurf1 affects the process of osteogenic differentiation through various mechanisms. Runt-related transcription factor 2 (Runx2) is a core transcription factor for directed differentiation of the osteoblast cell lineage and plays a crucial role in promoting osteoblast differentiation. It is involved in the Ub-proteasome pathway, which regulates osteoblast differentiation and bone formation by interacting with and ubiquitinating Runx2 (30). The BMP signaling pathway plays a critical biological role in bone development and post-natal bone formation, mainly by regulating the differentiation and function of mesenchymal stem cells (MSCs) and osteoblasts. Smurf1 degrades BMP downstream signaling molecules also in an Ub-dependent manner and when Smurf1 is inhibited, bone formation is enhanced in osteoporotic mice with markedly age-related osteoporosis (31). Jun B proto-oncogene (JunB) is an ubiquitinated substrate of Smurf1 and a stimulator of MSC proliferation and differentiation into osteoblasts. Smurf1 is able to bind to and ubiquitinate JunB, which is subsequently degraded by the proteasome. This degradation process reduces the protein level of JunB, which inhibits its transcriptional activity and further affects the proliferation and differentiation of MSCs, leading to the regulation of bone metabolism (32). Additionally, the P53/microRNA (miR/miRNA)-17/Smurf1 pathway inhibits osteogenesis through suppressing MSC function in age-related models, leading to osteoporosis (33). However, the underlying mechanism remains elusive.

Smurf2

Smurf1 and 2 share ~70% sequence homology and similar structural features (34), but Smurf2 has an additional WW structural domain, meaning that the functionality is different. Compared with Smurf1−/− mice, the osteoclast number in Smurf2−/− mice notably increased resulting in severe osteoporosis (35). Transforming growth factor-β (TGF-β) signaling is involved in the maintenance of bone homeostasis; TGF-β induces osteoblast differentiation and proliferation, inhibiting osteoclast formation (36). Smurf2 is transmitted primarily by TGF-β signaling, which is an important inhibitor of TGF-β. Smurf2 ubiquitinates the TGF-β receptor and Smad proteins to regulate the TGF-β signaling pathway and maintain tissue homeostasis (37). Furthermore, BMSCs from Smurf2−/− mice showed increased osteogenesis, enhancing the BMP/Smad signaling pathway. More specifically, Smad3 ubiquitination by Smurf2 negatively regulates BMP/Smad signaling (38). In the skeletal system, Smurf2 has been investigated and it was shown to play an important regulatory role in the transfer of information between osteoblasts and osteoclasts. More specifically, Smurf2 is able to regulate the activity of osteoclasts, which are dependent on osteoblasts. This means that Smurf2 does not act directly on osteoclasts, but indirectly regulates osteoclast behavior by affecting osteoblasts. This regulatory mechanism may involve signaling and interactions between osteoblasts and osteoclasts to maintain homeostasis in the skeletal system (35).

WW domain-containing E3 Ub protein ligase 1 (WWP1)

WWP1 is a 922-amino acid long HECT type E3 Ub protein ligase that interacts with the proline-rich polypeptide motif of target proteins to regulate protein transport, degradation, signaling and transcription. The role of WWP1 in bone homeostasis was first discovered in 2006 by Shu et al (39). WWP1 can polyubiquitinate Runx2 and decrease the differentiation and migration of osteoblasts. The role of the intracellular adaptor protein Schnurri-3 (Shn3) in osteoblastic bone formation is well established. Shn3 is able to regulate Runx2 protein levels through a delicate mechanism. Specifically, Shn-3 is able to interact with WWP1 and recruit it to Runx2. Once WWP1 binds to Runx2, it is able to promote the ubiquitination of Runx2, which in turn triggers the process of Runx2 degradation. In this way, Shn3 effectively controls the intracellular levels of the Runx2 protein, indirectly affecting the process of bone tissue formation and mineralization (40). Additionally, during inflammation-mediated osteoporosis models, WWP1 targets JunB for ubiquitination and degradation in MSCs to inhibit MSC differentiation into osteoblasts (41). As with WWP1, WWP2 belongs to the HECT type of E3 Ub protein ligases. WWP2 mediates monoubiquitination of Runx2 leading to its transactivation and resulting in increased osteogenic activity and promotion of osteogenic differentiation (42).

RNF146

The RING finger (RNF) 146, a RING domain and PAR-acylation dependent cytoplasmic E3 ligase, is indispensable in bone formation. RNF146 can improve MSC self-renewal and osteogenesis/adipogenesis, leading to increased bone mass (43). In osteoclast differentiation, RNF146 can promote the ubiquitination and degradation of AXIN, thereby regulating the Wnt signaling pathway and activating β-catenin to promote osteoclast differentiation. In addition, BP2 activity is indispensable for SRC tyrosine kinase in osteoclast differentiation. RNF146 deubiquitinates 3BP2, thereby inhibiting SRC activity and reducing osteoclast differentiation ability. Furthermore, RNF146 expression can be suppressed by RANKL through the NF-κB pathway, by regulating the stability of AXIN and 3BP2, thereby intervening in osteoclast differentiation (44).

RNF185

RNF185, also known as FLJ38628, is a RING domain E3 Ub ligase. In a high-throughput screening (45) of synthetic silencing RNA libraries targeting 5,000 human genes, RNF185 was identified as an endogenous inhibitor of osteoblastic norms. Deficiency of RNF185 can initiate human MSCs to differentiate into osteoblasts. An additional study further revealed the underlying mechanism through which RNF185 negatively regulates osteogenesis by degrading dishevelled (Dvl) 2 and downregulating the Wnt signaling pathway (46).

Itchy E3 ubiquitin protein ligase (ITCH)

In bone metabolism, ITCH negatively regulates the differentiation of mesenchymal progenitor cells into osteoblasts by mediating the proteasomal degradation of the JunB protein, a positive osteoblast regulator (47). In addition, in animal experiments, high expression of ITCH was detected in mouse fracture callus. Several positive regulatory factors for osteoblast differentiation, including Runx2, increase in the callus of Itch−/− mice fractures and thus the differentiation of osteoblasts is also markedly enhanced (48). In osteoclast formation, ITCH inhibits TRAF6 de-ubiquitination by binding the deubiquitinating enzyme cylindromatosis to inhibit RANKL-induced osteoclast. ITCH transcription is directly regulated by RANKL during osteoclast differentiation via the NF-κB (49).

Others

S-phase kinase-associated protein (SKP) 2 regulates a wide range of cell cycle proteins by targeting the SKP1-Cullin1-F-box for ubiquitination and proteasomal degradation (50). SKP2 negatively targets Runx2 for Ub-mediated degradation to inhibit osteogenic differentiation (51). Muscle-specific RING finger E3 ligase 1 (MURF1), a RING-type E3 Ub ligase, exhibits a critical role in inducing skeletal muscle atrophy. It has been reported that prolonged bed rest for various reasons, such as fractures, spinal cord injury, brain disease and exposure to microgravity, causes loss of muscle mass and strength, which further leads to bone loss. Mechanistically, MURF1 is upregulated and increases protein degradation and muscle wasting in numerous muscle atrophy models (52). The suppression of MURF1 can reduce osteoclastogenesis (53). Ubiquitin-conjugating enzyme E2 E3 (UBE2E3) is negatively and positively associated with age and osteogenic genes, respectively. The lack of UBE2E3 was further shown to promote osteoporosis development and overexpression could inhibit cell aging and increase osteogenic differentiation of old BMSCs (54). TRAF4 is markedly reduced in bone sections of ovariectomized rats with local osteoporosis and patients with osteoporosis (55). Further mechanisms indicate that the TRAF4 degrades Smurf2 to enhance osteoblastic differentiation of MSCs. Neural precursor cell expressed developmentally downregulated protein 4 (NEDD4), a member of the HECT domain family, participates in cell proliferation, apoptosis, autophagy and other biological processes to regulate mammalian growth and development (56,57). NEDD4 enhances osteoblast proliferation and bone mass accumulation by inducing the degradation of TGF-1β-activated pSMAD1 and improving the activation of the pSMAD2 and pERK1/2 pathways (58).

3. Role of DUBs in bone remodeling

DUBs are involved in DNA repair, gene transcription, apoptosis, autophagy, DNA repair and immune responses (19). DUBs act as critical regulators for bone remodeling by regulating basic multicellular unit differentiation and/or function.

Ub-specific proteases (USPs) and osteoblasts

Human adipose-derived stem cells (hASCs) can be extracted and isolated from human adipose tissue and have the potential for self-renewal and multidirectional differentiation. hASCs can be induced to differentiate in a variety of directions, such as adipogenic, osteogenic and chondrogenic. During osteogenic differentiation of hASCs, the expression level of USP7 changes and is positively associated with the degree of osteogenic differentiation. This implies that USP7 may play an important role in the osteogenic differentiation of hASCs (59). USP1 controls the NF-κB signaling pathway by inhibiting TRAF6 ubiquitination, thereby inhibiting osteoblast pyroptosis (60). USP1 deletion results in osteogenic impairment in mice and marked osteopenia. Overexpression of USP1 in MSCs inhibits osteogenic differentiation and promotes the preservation of MSC features. The mechanism is that USP1 deubiquitinated inhibitors of DNA-binding proteins to promote the preservation of MSC signatures (61). USP8 in bone progenitor cells prevents the degradation of the Frizzy-5 ubiquitinated Wnt receptor to ensure Wnt-induced osteogenesis, thus promoting bone formation (62). USP11 can stabilize the Msh homeobox 1 (MSX1) protein to upregulate the expression of osteogenic differentiation-related markers in human MSCs, consequently enhancing osteogenic differentiation (63). The USP34 protein is involved in bone formation by modulating BMP2 signaling in MSCs. When BMSCs or pre-osteoblasts are conditionally knocked out of USP34, mice have low bone mass. Mechanically, USP34 stabilizes both Smad1 and RUNX2 for osteogenic differentiation and bone formation (64). Ubiquitin C-terminal hydrolase-L3 (UCHL3) regulates BMP2-induced Smad1 polyubiquitination to promote osteoblast differentiation (65).

USP4

USP4 affects osteogenic differentiation through multiple mechanisms. The ubiquitination of the Dvl protein can activate the Wnt/β-catenin signaling pathway to affect osteoblast differentiation (66). By removing the lysine-63-linked polyUb chain from Dvl, USP4 strongly inhibits Wnt/β-catenin signal transduction to weaken osteogenic differentiation (67). Regulatory pathways associated with TGF/BMP are also important in bone growth (68). USP4 can target the TGF-β receptor for deubiquitination and enhance the activation of the TGF-β/BMP signaling pathway to promote the proliferation and differentiation of MSCs (69).

USP53

USP53 enhances osteogenic function and differentiation in human BMSCs by stabilizing key proteins associated with osteogenic differentiation and regulating the BMP signaling pathway. Moreover, USP53-overexpressing human BMSCs markedly promote bone regeneration in murine calvaria bone defects. Mechanistically, the interaction of USP53 with F-box protein 31 revealed a new role for USP53 in cellular signaling and protein degradation. This interaction specifically involves the degradation of β-catenin by the Skp1/Cul1/F-box protein complex, a key process that is essential for the regulation of the Wnt signaling pathway (70). In addition, USP53 acting on the vitamin D-receptor-SMAD3 pathway increases RANKL-dependent osteoclastogenesis through osteoblasts (71).

UCHL1

UCHL1 was originally identified in neurons. CXCL7, a chemokine with a C-X-C motif, is associated with the posterior longitudinal ligament ossification of the spine (OPLL) phenotype, manifested by dyskinesia, ectopic ossification of posterior ligament tissue and osteoporosis. UCHL1 is overexpressed in the serum of mice with a CXCL7 gene deletion and in patients with OPLL (72). In patients with periodontitis, UCHL1 leads to periodontal ligament stem cell osteogenesis and alveolar bone loss by regulating the BMP2/Smad signaling pathway. However, it is unclear which molecule is deubiquitinated by UCHL1. By inhibiting the high expression of UCHL1, periodontal bone loss is reduced and inflammation is alleviated (73).

USPs and osteoclasts
Cylindromatosis (CYLD)

CYLD negatively regulates osteoclastogenesis. Consistently, the number and activity of osteoclasts in CYLD knockout mice increases and the mice exhibit severe osteoporosis (74). With the use of proteasome inhibitors, CYLD accumulation increases, leading to impairment of osteoclast production and function (75). Mechanically, CYLD, through deubiquitination, is able to remove the Ub chain on TRAF6 and inactivate it, thereby inhibiting the activation of the RANK signaling pathway (76).

A20

A20 has been shown to mainly regulate osteoclast formation. A20 overexpression has anti-inflammatory effects on human periodontal ligament cells (HPDLCs) and blocks osteoclast differentiation (77). A20 knockout mice have increased osteoclasts, thinner bone trabeculae and are prone to develop severe osteoporosis (78). A20 induction by intravenous gamma globulin attenuates RANKL inducing NF-κB signaling to reduce osteoclastogenesis in vivo as well as inhibits bone resorption, thus preserving bone mass (79). Mechanistically, the A20 anti-osteoclast function is mainly dependent on inhibiting NF-κB activation by altering IκBα ubiquitination-mediated degradation, which physically interacts with NF-κB to inhibit its activation (80). TRAF6-dependent autophagy in HPDLCs is inhibited for the reduced expression of A20 with 2% hypoxia treatment, resulting in decreased osteoclast differentiation and formation. The underlying mechanism involves A20 inhibiting K63-linked, while it enhances K48-based ubiquitination of TRAF6 to suppress TRAF6 activation and promote TRAF6 proteasomal degradation, respectively, thus inhibiting autophagy under hypoxic conditions (81).

Jab1/Mpr1/Pad1 N-termina domain associated metalloisopeptidase (JAMM/MPN)

Myb-like, Swi3p, Rsc8p and Moira (SWIRM) and MPN domain containing 1 (MYSM1) is a metalloprotease composed of three domains, MPN, SWI2/SNF2 ISWI-like SANT (SANT) and SWIRM (82). MYSM1 is not only an important regulator of hematopoiesis and immunity (83), but also essential for maintenance of bone homeostasis. Mysm1−/− mice showed enhanced autonomic differentiation of MSCs and accelerated adipogenesis, as evidenced by reduced bone mass in long bones and cranial bones (84). A study demonstrated that osteoclast progenitor cells in Mysm1−/− mice showed decreased proliferative ability and markedly decreased osteoclast number and absorption activity (85). However, the underlying mechanism of MYSM1 in osteoclast differentiation and function remains enigmatic.

Others

USP7 has a dual effect on osteoclasts. On the one hand, in CD14+ human peripheral blood mononuclear cell studies, USP7 promotes osteoclast differentiation through high mobility group box 1 (HMGB1) deubiquitination (86). On the other hand, USP7 negatively regulates osteoclastogenesis through a mixed mechanism of deubiquitination-mediated TRAF6 signaling and deubiquitination-mediated degradation of stimulator of interferon genes (STING) signaling (87). USP18 specifically removes the ISGylation from substrate proteins by interferon-stimulated (ISG) gene 15 and also regulates the interferon signaling pathway to block ISGs (88,89). The differentiation ability and bone resorption activity are increased in USP18-deficient mice, resulting in an osteoporotic phenotype in mice (90). USP34 is not only required for osteogenic differentiation, but also inhibits osteoclastogenesis. USP34 deficiency leads to RANKL-mediated NF-κB activation and increased osteoclast production, which in turn leads to a phenotype of bone loss in mice. The specific process of this mechanism is as follows: Under normal conditions, USP34 inhibits the activation of the NF-κB pathway by stabilizing IκBα, a protein that inhibits NF-κB, through deubiquitination. However, in the absence of USP34, the stability of IκBα is reduced, resulting in excessive activation of the NF-κB pathway. This activated state enables RANKL to bind easily to its receptor RANK, which in turn promotes osteoclast generation and bone resorption function (91). USP26 plays a balancing role during osteogenic and osteoclast differentiation. USP26 enhances IκBα stability to inhibit NF-κB activation, consequently suppressing osteoclast differentiation in bone marrow-derived macrophages (92). Moreover, in bone defect mice, USP26 increases osteogenic differentiation, promotes bone regeneration and reduces bone loss (92). OTU domain-containing Ub aldehyde-binding protein 1 in osteoblasts antagonizes the ubiquitination of Fibroblast Growth Factor Receptor 2 (FGFR2) mediated by the Ub ligase SMURF1 in an enzyme activity-independent manner to maintain protein stability, resulting in enhanced osteoblast formation (93).

4. Medication for osteoporosis

The main goal of osteoporosis medications is to reduce the risk of osteoporotic fractures and improve the quality of life of patients by relieving pain, increasing bone density and preventing fractures (94). According to the different mechanisms of action, anti-osteoporosis drugs are generally placed in three categories: i) Inhibitors of bone resorption; ii) promoters of bone formation; and iii) dual-acting drugs. Bone formation promoters are mainly parathyroid hormone receptor agonists, including teriparatide and appalatide. Sclerostatin monoclonal is a double-acting drug (95). There are five main categories of bone resorption inhibitors: i) Bisphosphonates (BPs); ii) RANKL inhibitors; iii) estrogen; iv) selective estrogen receptor modulators; and iv) calcitonin (Table II).

Table II

Common anti osteoporosis medication guideline recommendations and precautions.

Table II

Common anti osteoporosis medication guideline recommendations and precautions.

A, Bone resorption inhibitors
Action mechanismDrug typeDrug namesGuideline recommendationAdverse reactionsContraindications and important warnings
BisphosphonatesAlendronate, Isedronate, ibandronate, zoledronic acidThe first-line option and the most cost-effective interventions. Effectively reduce the risk of multiple fractures (vertebral, non-vertebral and hip)Oral: Astrointestinal symptoms such as abdominal pain, diarrhea, nausea, dyspepsia and constipation; Intravenous: Influenza-like symptoms occur in 1/3 of patients within 3 days of medicationZoledronic acid is prohibited (creatinine clearance below 35 ml/min). Preoperative improvement of oral problems may reduce the risk of osteonecrosis of the jaw. Oral for 5 years and intravenous for 3 years, re-evaluate and consider the drug holiday.
RANK ligand inhibitorDenosumabEffectively reduces the risk of multiple fractures (vertebral, non-vertebral and hip) (96). Approved for use only in postmenopausal women in some countries.Hycalcemia, severe infection (cystitis, respiratory tract infection, skin cellulitis, etc.), rash, skin pruritus, myalgia or bone pain, etc.; individual reports of jaw necrosis and atypical femur fracture.Ensure that the patient knows the osteoporosis management plan.
Desumamab cannot be easily stopped to avoid rapid decline of bone mineral density
EstrogenEstradiol, estropipate, conjugated oestrogenLow-risk postmenopausal womenHeadache, edema, tumor, thrombusIncreased risk of cardiovascular events and breast cancer. Initiation of treatment for more than 10 years after menopause is not recommended;
Selective estrogen receptor modulatorsRaloxifene bazedoxifene bazedoxifene and conjugated oestrogenReduces vertebral risk in postmenopausal osteoporotic womenVasomotor symptoms, muscle spasm. Occasional venous thrombosisVenous thromboembolism. Increased risk of stroke death in women at high risk of coronary heart disease. For women only.
Calcitonin(Human, salmon)Osteoporosis and its inducedFace flushing, nausea, allergy bone painNote the allergy test

B, Bone formation promoters
Parathyroid hormone receptor agonistTeriparatide and appalatideTeriparatide is the first drug for postmenopausal osteoporosis, male and glucocorticoid-induced osteoporosis with high risk of fracture (97)Nausea, limb pain, headache and vertigoThe duration of treatment should not exceed 24 months. Theoretically there is an increased risk of osteosarcoma (91). Contraindicated for patients with prior bone radiotherapy, bone malignancies, bone metastases or hypercalcemia

C, Dual-acting drugs
Sclerostin monoclonal antibodyRomosozumabFirst-line treatment for women with high risk of postmenopausal fracture, especially for patients with spinal fractureArthralgia, nasopharyngitis, back pain and injection site reactions, rare mandibular osteonecrosis and atypical fracturesThe duration of treatment should not exceed 24 months. Be aware of potential cardiovascular risks

[i] RANK, receptor activator of nuclear factor-κB.

BPs are widely used in the treatment of osteoporosis, where they are effective in inhibiting bone resorption and increasing bone mass at vertebral, non-vertebral and hip sites, thereby reducing the risk of fracture (96). However, oral BPs can increase upper gastrointestinal symptoms in patients and ~33% of patients receiving first intravenous zoledronic acid develop flu-like symptoms. In addition, prolonged use of BP medications may result in reduced blood supply to the jaws, thereby increasing the risk of osteonecrosis of the jaws as well as inhibition of bone turnover, leading to impaired bone remodeling, thereby increasing the risk of atypical fractures (97). Denosumab is also the most common osteoporosis drug in the clinic. It can be combined with RANKL to markedly reduce the degree of osteoporosis in patients. If Denosumab was stopped without other osteoporosis drugs, bone mineral density showed a steep decline (98). It is noteworthy that rashes, cellulitis, femoral shaft or subtrochanteric fractures and jaw necrosis may occur during use. The application of estrogen in women aged <60 years or 10 years after menopause can effectively prevent bone loss caused by menopause. Estrogen acts by indirectly regulating calcitonin, parathyroid hormone and 1,25-(OH)2D3. However, Estrogen replacement therapy has been limited clinically due to potential adverse effects, including venous thromboembolism, cerebrovascular disease, stroke and breast cancer (99). Menopausal hormone therapy (MHT) has its unique advantages in the treatment of postmenopausal osteoporosis (100,101). In the mid-1990s and early 2000s, in most European and North American countries, MHT is widely used to prevent the risk of postmenopausal osteoporosis and related bone fractures in women. Following the publication of the randomized Women's Health Initiative (WHI) report (102), which found that MHT increased the risk of coronary heart disease, stroke and breast cancer, there was a dramatic decline in MHT prescribing worldwide. The current trend is to recommend lower doses of estrogen for effective relief of vasomotor symptoms and prevent vaginal atrophy with an improved bleeding profile than with higher estrogen dosages within 10 years of menopause than was recommended a few years ago. For the prevention of endometrial cancer caused by MHT, it is recommended to add progesterone (103). In addition, compared with oral estrogen therapy, transdermal estrogen patch can reduce the risk of venous thrombosis and atherosclerotic vascular disease and also protect the heart (104). Therefore, low-dose non-oral MHT may be considered for women with cardiovascular risk factors, older women and those who choose to use MHT for extended periods to protect bone (105,106). In conclusion, individualized treatment is advocated for hormone replacement therapy and different formulations, dosages and MHT regimens are considered at the same time. The artificial synthesis of selective estrogen receptor modulators can selectively act on different tissue estrogen receptors to reduce the incidence of breast cancer in women (107). However, some side effects, including vasomotor symptoms, muscle spasm and venous thrombosis, remain. The Use of Raloxifene in Heart Disease (RUTH) study showed that Raloxifene increased fatal stroke (0.7‰ increase in absolute risk) and the risk of the occurrence of venous thromboembolic disease (absolute risk increase by 1.3‰) (108).

Parathyroid hormone is one of the important hormones that regulate calcium and phosphorus metabolism, as well as bone conversion. It has been shown that parathyroid hormone promotes bone anabolic metabolism that is dependent on low-dose and intermittent administration. The duration and dosage of teriparatide use are closely associated with rat bone tumors (109), but post-marketing surveillance did not reveal a higher than expected risk of osteosarcoma (110). Calcitonin reduces blood calcium and phosphorus mainly by inhibiting osteoclasts. However, long-term use of calcitonin leads to clinical resistance. In addition, there may be an increased risk of cardiovascular complications. This may be associated with the effect of calcitonin on the cardiovascular system; it may affect the systolic and diastolic function of the heart, or the relaxation and contraction of blood vessels (111). Monoclonal sclerostin romosozumab promotes the formation of bone mass, while it inhibits the resorption of bone, rapidly forming bone on trabecular and cortical bone surfaces to increase bone density and strength (112). The most common adverse reactions include arthralgia, nasopharyngitis, back pain and injection site reactions, rare mandibular osteonecrosis and atypical fractures. The black box warning on the potential cardiovascular risks of romosozumab, including serious adverse events such as non-fatal myocardial infarction, non-fatal stroke and cardiovascular death, may indeed place some limitations on its clinical use (95). Osteoporosis guidelines emphasize vitamin D and calcium supplementation as basic measures. Importantly, it has been shown that when patients with osteoporosis are treated with anti-osteoporosis drugs, the effectiveness of these drugs is markedly reduced without adequate vitamin D and calcium supplementation (113).

5. Clinical application and prospects of UPS

UPS is critical for bone cell formation and differentiation. Based on these mechanisms of action, drugs targeting these Ub ligases and deubiquitinating enzymes for the treatment of osteoporosis and other related bone diseases continue to be developed. For example, by inhibiting the activity of certain Ub ligases, it may be possible to promote bone formation or inhibit bone resorption for the treatment of osteoporosis. By contrast, by activating the activity of certain deubiquitinating enzymes, it may also be possible to have a positive impact on the treatment of skeletal diseases.

Catalpol, a biologically active cyclic enol ether terpene glucoside extracted from the traditional Chinese Medicine Rehmannia glutinosa, inhibits the ubiquitination and degradation of phosphatase and tensin homolog to increase phosphatase activity and then inhibits downstream AKT and NF-κB associated with RANKL signal pathways, consequently suppressing osteoclast formation and bone absorption (114). In mice with LPS and ovariectomized-induced bone loss, catalpol improves bone loss by attenuating osteoclast activity (114). Melatonin is a biologically active amine hormone secreted by the pineal gland of the brain and is a signaling molecule with a circadian rhythm, participating in various physiological processes including bone metabolism (109). Melatonin inhibits RANKL receptor activators to inhibit osteolysis and prevent bone loss and also promotes osteoblast differentiation and activity by melatonin receptor 2 (115). Melatonin reduces the ubiquitination of the Smad1 protein preventing its degradation by inhibiting Smurf1 activity, thereby stabilizing BMP Smad1 signaling activity and restoring inflammatory factor-induced osteogenesis (116). Carnosic acid (CA), an amino acid substance with strong antioxidant and anti-aging effects, is widely used in weight loss foods, nutritional supplements and other fields. CA binds to the ligand-binding region of cholesterol and estrogen receptor-associated receptor α, promoting its ubiquitination and proteasomal degradation, thereby inhibiting RANKL-associated osteoclast formation and ovariectomy-induced bone loss (117).

Beraprost can improve postmenopausal osteoporosis by downregulating the NEDD4-mediated ubiquitination-proteasome degradation of Runx2 (118). The selective complexes B06 and B75 block binding of Smurf1 to Ub, thereby stabilizing the level of the BMP signaling component Smad1/5 protein and enhancing osteoblast activity. This also offers promising new clinical prospects for the therapy of osteoporosis (119). miR-195-5p can target and inhibit the activation of BMP-2/SMAD/Akt/Runx2 pathway signaling by Smurf1, to promote osteogenic differentiation in ovariectomized mouse models (120). Chalcone derivatives enhance local bone mass after lumbar spine fusion surgery in normal BMP-2 mice with high expression of Smurf1 (31). Bortezomib, is a 26S proteasome inhibitor which inhibits the degradation of polyUb protein by suppressing the hydrolysis activity of the proteasome. Bortezomib regulates proteasomal degradation of Runx2 via the Wnt/β-catenin signaling pathway (121), inducing osteogenic differentiation and promoting osteogenesis (122). Furthermore, a recent study suggested that bortezomib may treat post-menopausal osteoporosis by inhibiting the Smurf-mediated ubiquitination-proteasome process (123). Withaferin A, also a proteasome inhibitor, promotes osteoblast proliferation and differentiation as well as regulates bone anabolism to promote osteoporotic fracture healing (124).

miRNAs participate in the progress in osteoporosis through a complex signaling pathway network (125). miR-21-5p inhibits osteoclast differentiation by targeting SKP2. In an ovariectomy (OVX) mouse model injected with miR-21-5p, the number of osteoclasts was reduced and the degree of osteoporosis was improved (126). Neat1, the mechanoreceptor long non-coding RNA (lncRNA), under mechanical stimulation, regulates osteogenic function by nuclear retention of the paraplaque-dependent E3 Ub ligase Smurf1 mRNA. In Neat1-knockout hindlimb unloading mice, no bone damage or bone loss are found. This provides a treatment reference for bone loss caused by long-term space flight or inactivity and osteoporosis associated with aging (127). In addition, SNHG1 lncRNA inhibits osteogenic differentiation through NEDD4-mediated ubiquitination by suppressing the activation of the p38-MAPK signaling pathway, a key trigger factor in bone formation (128). In recent years, a new drug delivery system, nanoparticles, has been used to treat osteoporosis. Ferumoxytol and ferucarbotran, as clinical nanoparticles, exert anti-osteoporosis effects by inhibiting osteoclastogenesis and participating in bone metabolism. Nanoparticles trigger the upregulation of p62, resulting in the recruitment of CYLD to enhance deubiquitination and inactivation of TRAF6, the main controller of the RANKL and NF-κB signal (129). When downstream activation of the NF-κB signaling pathway and the MAPK signaling pathway is attenuated, the proliferation and differentiation processes of osteoclast precursors may be impeded, leading to a reduction in osteoclast formation. Intravenous injection can improve bone quality in OVX mice and it is anticipated to be an alternative drug for the treatment of osteoporosis (129).

In addition, several clinically used drugs have been discovered to be involved in bone homeostasis via the UPS system, such as thalidomide (130), lansoprazole (131), vitisin A, clomimidazole and zoledronic acid (132), all of which inhibit or induce ubiquitination of related substrates.

6. Conclusion and future perspectives

The present review introduced and summarized the current literature on ubiquitination and deubiquitination in osteoporosis and bone homeostasis, highlighting the critical importance of Ub-dependent proteolytic systems in skeletal cell function. The E3 connecting enzymes are mainly regulated by transcription factors and they have notable protein participation in bone formation in the signaling pathway of BMSC and osteoblast proliferation and differentiation. Only a few studies have shown that RNF146 (44) and Smurf2 (35) regulate osteoclasts. The E3 protease system can influence the bone remodeling process by regulating different target proteins. They recognize and degrade proteins that are involved in bone formation, thereby regulating the rate and extent of bone formation. At the same time, they also recognize and degrade proteins involved in bone resorption, thereby controlling the process of bone resorption. This precise regulatory mechanism is essential for maintaining bone homeostasis.

Research on the regulation of deubiquitinating enzymes on osteoblasts and osteoclasts is limited and the mechanism and molecules of action have not been fully elucidated. To date, the relationship between MJDs and MINDY families with skeletal cell differentiation and function has not been reported. Elucidating the intrinsic mechanisms by which DUB gene alterations regulate osteoblast and osteoclast functions is important for the development of novel and more effective therapeutic strategies for osteoporosis. Designing drugs targeting specific genetic alterations in the DUB gene to avoid its dysregulation is one of the important directions for developing novel osteoporosis treatment strategies. Through in-depth study of the function and regulatory mechanisms of the DUB gene, potential drug targets can be identified and targeted drugs can be designed to regulate the function of osteoblasts and osteoclasts. These drugs can correct the imbalance of bone metabolism by altering the activity or expression level of the DUB gene, thus achieving the goal of treating bone diseases such as osteoporosis. In the future, through in-depth study of the function and regulatory mechanism of the DUB gene, as well as the design of drugs targeting its specific genetic alterations, it is anticipated to provide new ideas and methods for the treatment of skeletal diseases such as osteoporosis.

Meanwhile, several anti-osteoporosis drugs commonly used in the clinic were discussed in the present review as well as their disadvantages and the current clinical studies on ubiquitination-related drugs for osteoporosis. At present, anti-osteoporosis drugs have shown some efficacy in clinical research and their benefits outweigh the risk of side effects, but patients lack compliance due to the aforementioned side effects, high costs and sequential treatment. Although designing drugs to target specific genetic alterations in the DUB gene is challenging and the adverse effects and the interactions with other drugs are not yet clear, the future research direction may be to treat osteoporosis by mediating ubiquitination.

Furthermore, the internal mechanisms by which ubiquitination regulates skeletal cell function have not been thoroughly investigated and the specific sites of ubiquitination modification and the interactions of different molecules acting at the same site have not been clarified. Decoding the pathways between ubiquitination and other modification mechanisms has also not been described in depth. These practical mechanisms should be further investigated in the future.

Availability of data and materials

Not applicable.

Authors' contributions

CC conceived the study. XF wrote the first draft. RZ drew the figures. GX, WL, PF, CC and RG revised the manuscript and figures. All authors read and approved the final 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.

Acknowledgements

Not applicable.

Funding

The present review was supported by grants from the Qinghai Provincial Department of Science and Technology (grant no. 2022-ZJ-951Q).

References

1 

The Lancet Diabetes Endocrinology: Osteoporosis: Overlooked in men for too long. Lancet Diabetes Endocrinol. 9:12021. View Article : Google Scholar

2 

Shen Y, Huang X, Wu J, Lin X, Zhou X, Zhu Z, Pan X, Xu J, Qiao J, Zhang T, et al: The global burden of osteoporosis, low bone mass, and its related fracture in 204 countries and territories, 1990-2019. Front Endocrinol (Lausanne). 13:8822412022. View Article : Google Scholar : PubMed/NCBI

3 

Zhu Y, Huang Z, Wang Y, Xu W, Chen H, Xu J, Luo S, Zhang Y, Zhao D and Hu J: The efficacy and safety of denosumab in postmenopausal women with osteoporosis previously treated with bisphosphonates: A review. J Orthop Translat. 22:7–13. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Xu XM, Li N, Li K, Li XY, Zhang P, Xuan YJ and Cheng XG: Discordance in diagnosis of osteoporosis by quantitative computed tomography and dual-energy X-ray absorptiometry in Chinese elderly men. J Orthop Translat. 18:59–64. 2018. View Article : Google Scholar

5 

Zhang YW, Cao MM, Li YJ, Dai GC, Lu PP, Zhang M, Bai LY, Chen XX, Zhang C, Shi L and Rui YF: The regulative effect and repercussion of probiotics and prebiotics on osteoporosis: Involvement of brain-gut-bone axis. Crit Rev Food Sci Nutr. 63:7510–7528. 2023. View Article : Google Scholar

6 

Intemann J, De Gorter DJJ, Naylor AJ, Dankbar B and Wehmeyer C: Importance of osteocyte-mediated regulation of bone remodelling in inflammatory bone disease. Swiss Med Wkly. 150:w201872020.PubMed/NCBI

7 

Amarasekara DS, Kim S and Rho J: Regulation of osteoblast differentiation by cytokine networks. Int J Mol Sci. 22:28512021. View Article : Google Scholar : PubMed/NCBI

8 

Edwards JR and Mundy GR: Advances in osteoclast biology: Old findings and new insights from mouse models. Nat Rev Rheumatol. 7:235–243. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Wu M, Chen G and Li YP: TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 4:160092016. View Article : Google Scholar

10 

Han L, Wu J, Wang M, Zhang Z, Hua D, Lei S and Mo X: RNA modification-related genetic variants in genomic loci associated with bone mineral density and fracture. Genes (Basel). 13:18922022. View Article : Google Scholar : PubMed/NCBI

11 

Wang C, Chen Q and Xu H: Wnt/β-catenin signal transduction pathway in prostate cancer and associated drug resistance. Discov Oncol. 12:402021. View Article : Google Scholar

12 

Hu R, Chen L, Chen X, Xie Z, Xia C and Chen Y: Aloperine improves osteoporosis in ovariectomized mice by inhibiting RANKL-induced NF-κB, ERK and JNK approaches. Int Immunopharmacol. 97:1077202021. View Article : Google Scholar

13 

Hou H, Peng Q, Wang S, Zhang Y, Cao J, Deng Y, Wang Y, Sun WC and Wang HB: Anemonin attenuates RANKL-induced osteoclastogenesis and ameliorates LPS-induced inflammatory bone loss in mice via modulation of NFATc1. Front Pharmacol. 10:16962020. View Article : Google Scholar : PubMed/NCBI

14 

Deng L, Meng T, Chen L, Wei W and Wang P: The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduct Target Ther. 5:112020. View Article : Google Scholar : PubMed/NCBI

15 

Cai J, Culley MK, Zhao Y and Zhao J: The role of ubiquitination and deubiquitination in the regulation of cell junctions. Protein Cell. 9:754–69. 2018. View Article : Google Scholar :

16 

van Huizen M and Kikkert M: The role of atypical ubiquitin chains in the regulation of the antiviral innate immune response. Front Cell Dev Biol. 7:3922020. View Article : Google Scholar : PubMed/NCBI

17 

Akutsu M, Dikic I and Bremm A: Ubiquitin chain diversity at a glance. J Cell Sci. 129:875–880. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Gundogdu M and Walden H: Structural basis of generic versus specific E2-RING E3 interactions in protein ubiquitination. Protein Sci. 28:1758–1770. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Mennerich D, Kubaichuk K and Kietzmann T: DUBs, hypoxia, and cancer. Trends Cancer. 5:632–653. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Komander D and Rape M: The ubiquitin code. Annu Rev Biochem. 81:203–229. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zheng N and Shabek N: Ubiquitin ligases: Structure, function, and regulation. Annu Rev Biochem. 86:129–157. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Zhao J, Guo J, Wang Y, Ma Q, Shi Y, Cheng F, Lu Q, Fu W, Ouyang G, Zhang J, et al: Research progress of DUB enzyme in hepatocellular carcinoma. Front Oncol. 12:9202872022. View Article : Google Scholar : PubMed/NCBI

23 

Clague MJ, Urbé S and Komander D: Breaking the chains: Deubiquitylating enzyme specificity begets function. Nat Rev Mol Cell Biol. 20:338–352. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Bello AI, Goswami R, Brown SL, Costanzo K, Shores T, Allan S, Odah R and Mohan RD: Deubiquitinases in neurodegeneration. Cells. 11:5562022. View Article : Google Scholar : PubMed/NCBI

25 

Hu M, Li P, Li M, Li W, Yao T, Wu JW, Gu W, Cohen RE and Shi Y: Crystal structure of a UBP-family deubiquitinating enzyme in isolation and in complex with ubiquitin aldehyde. Cell. 111:1041–1054. 2002. View Article : Google Scholar

26 

Johnston SC, Larsen CN, Cook WJ, Wilkinson KD and Hill CP: Crystal structure of a deubiquitinating enzyme (human UCH-L3) at 1.8 A resolution. EMBO J. 16:3787–3796. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Li Y and Reverter D: Molecular mechanisms of DUBs regulation in signaling and disease. Int J Mol Sci. 22:9862021. View Article : Google Scholar : PubMed/NCBI

28 

Komander D, Clague MJ and Urbé S: Breaking the chains: Structure and function of the deubiquitinases. Nat Rev Mol Cell Biol. 10:550–563. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Al-Rawi R, Al-Beshri A, Mikhail FM and McCormick K: Fragile bones secondary to SMURF1 gene duplication. Calcif Tissue Int. 106:567–573. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Glimcher LH, Jones DC and Wein MN: Control of postnatal bone mass by the zinc finger adapter protein Schnurri-3. Ann N Y Acad Sci. 1116:174–181. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Liang C, Peng S, Li J, Lu J, Guan D, Jiang F, Lu C, Li F, He X, Zhu H, et al: Inhibition of osteoblastic Smurf1 promotes bone formation in mouse models of distinctive age-related osteoporosis. Nat Commun. 9:34282018. View Article : Google Scholar : PubMed/NCBI

32 

Zhao L, Huang J, Guo R, Wang Y, Chen D and Xing L: Smurf1 inhibits mesenchymal stem cell proliferation and differentiation into osteoblasts through JunB degradation. J Bone Miner Res. 25:1246–1256. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Liu W, Qi M, Konermann A, Zhang L, Jin F and Jin Y: The p53/miR-17/Smurf1 pathway mediates skeletal deformities in an age-related model via inhibiting the function of mesenchymal stem cells. Aging (Albany NY). 7:205–218. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Koganti P, Levy-Cohen G and Blank M: smurfs in protein homeostasis, signaling, and cancer. Front Oncol. 8:2952018. View Article : Google Scholar : PubMed/NCBI

35 

Xu Z, Greenblatt MB, Yan G, Feng H, Sun J, Lotinun S, Brady N, Baron R, Glimcher LH and Zou W: SMURF2 regulates bone homeostasis by disrupting SMAD3 interaction with vitamin D receptor in osteoblasts. Nat Commun. 8:145702017. View Article : Google Scholar : PubMed/NCBI

36 

Bonewald LF and Mundy GR: Role of transforming growth factor-beta in bone remodeling. Clin Orthop Relat Res. 261–276. 1990.PubMed/NCBI

37 

Bai Y and Ying Y: The post-translational modifications of Smurf2 in TGF-β signaling. Front Mol Biosci. 7:1282020. View Article : Google Scholar

38 

Kushioka J, Kaito T, Okada R, Ishiguro H, Bal Z, Kodama J, Chijimatsu R, Pye M, Narimatsu M, Wrana JL, et al: A novel negative regulatory mechanism of Smurf2 in BMP/Smad signaling in bone. Bone Res. 8:412020. View Article : Google Scholar : PubMed/NCBI

39 

Shu L, Zhang H, Boyce BF and Xing L: Ubiquitin E3 ligase Wwp1 negatively regulates osteoblast function by inhibiting osteoblast differentiation and migration. J Bone Miner Res. 28:1925–1935. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Jones DC, Wein MN, Oukka M, Hofstaetter JG, Glimcher MJ and Glimcher LH: Regulation of adult bone mass by the zinc finger adapter protein Schnurri-3. Science. 312:1223–1227. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Zhao L, Huang J, Zhang H, Wang Y, Matesic LE, Takahata M, Awad H, Chen D and Xing L: Tumor necrosis factor inhibits mesenchymal stem cell differentiation into osteoblasts via the ubiquitin E3 ligase Wwp1. Stem Cells. 29:1601–1610. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Zhu W, He X, Hua Y, Li Q, Wang J and Gan X: The E3 ubiquitin ligase WWP2 facilitates RUNX2 protein transactivation in a mono-ubiquitination manner during osteogenic differentiation. J Biol Chem. 292:11178–11188. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Liu D, Kou X, Chen C, Liu S, Liu Y, Yu W, Yu T, Yang R, Wang R, Zhou Y and Shi S: Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res. 28:918–933. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Matsumoto Y, Larose J, Kent OA, Lim M, Changoor A, Zhang L, Storozhuk Y, Mao X, Grynpas MD, Cong F and Rottapel R: RANKL coordinates multiple osteoclastogenic pathways by regulating expression of ubiquitin ligase RNF146. J Clin Invest. 127:1303–1315. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Zhao Y and Ding S: A high-throughput siRNA library screen identifies osteogenic suppressors in human mesenchymal stem cells. Proc Natl Acad Sci USA. 104:9673–9678. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Zhou Y, Shang H, Zhang C, Liu Y, Zhao Y, Shuang F, Zhong H, Tang J and Hou S: The E3 ligase RNF185 negatively regulates osteogenic differentiation by targeting Dvl2 for degradation. Biochem Biophys Res Commun. 447:431–436. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Liu J, Li X, Zhang H, Gu R, Wang Z, Gao Z and Xing L: Ubiquitin E3 ligase Itch negatively regulates osteoblast function by promoting proteasome degradation of osteogenic proteins. Bone Joint Res. 6:154–161. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Zhang H and Xing L: Ubiquitin e3 ligase itch negatively regulates osteoblast differentiation from mesenchymal progenitor cells. Stem Cells. 31:1574–1583. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Zhang H, Wu C, Matesic LE, Li X, Wang Z, Boyce BF and Xing L: Ubiquitin E3 ligase Itch negatively regulates osteoclast formation by promoting deubiquitination of tumor necrosis factor (TNF) receptor-associated factor 6. J Biol Chem. 288:22359–22368. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Zhang F, Zhao Y and Sun Y: USP2 is an SKP2 deubiquitylase that stabilizes both SKP2 and its substrates. J Biol Chem. 297:1011092021. View Article : Google Scholar : PubMed/NCBI

51 

Thacker G, Kumar Y, Khan MP, Shukla N, Kapoor I, Kanaujiya JK, Lochab S, Ahmed S, Sanyal S, Chattopadhyay N and Trivedi AK: Skp2 inhibits osteogenesis by promoting ubiquitin-proteasome degradation of Runx2. Biochim Biophys Acta. 1863:510–519. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Eddins MJ, Marblestone JG, Suresh Kumar KG, Leach CA, Sterner DE, Mattern MR and Nicholson B: Targeting the ubiquitin E3 ligase MuRF1 to inhibit muscle atrophy. Cell Biochem Biophys. 60:113–118. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Bettis T, Kim BJ and Hamrick MW: Impact of muscle atrophy on bone metabolism and bone strength: Implications for muscle-bone crosstalk with aging and disuse. Osteoporos Int. 29:1713–1720. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Liu Y, Cai G, Chen P, Jiang T and Xia Z: UBE2E3 regulates cellular senescence and osteogenic differentiation of BMSCs during aging. PeerJ. 9:e122532021. View Article : Google Scholar : PubMed/NCBI

55 

Li J, Wang P, Xie Z, Wang S, Cen S, Li M, Liu W, Tang S, Ye G, Zheng G, et al: TRAF4 positively regulates the osteogenic differentiation of mesenchymal stem cells by acting as an E3 ubiquitin ligase to degrade Smurf2. Cell Death Differ. 26:2652–2666. 2019. View Article : Google Scholar : PubMed/NCBI

56 

An H, Krist DT and Statsyuk AV: Crosstalk between kinases and Nedd4 family ubiquitin ligases. Mol Biosyst. 10:1643–1657. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Wiszniak S, Harvey N and Schwarz Q: Cell autonomous roles of Nedd4 in craniofacial bone formation. Dev Biol. 410:98–107. 2016. View Article : Google Scholar

58 

Jeon SA, Lee JH, Kim DW and Cho JY: E3-ubiquitin ligase NEDD4 enhances bone formation by removing TGFβ1-induced pSMAD1 in immature osteoblast. Bone. 116:248–258. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Tang Y, Lv L, Li W, Zhang X, Jiang Y, Ge W and Zhou Y: Protein deubiquitinase USP7 is required for osteogenic differentiation of human adipose-derived stem cells. Stem Cell Res Ther. 8:1862017. View Article : Google Scholar : PubMed/NCBI

60 

Sun D, Peng Y, Ge S and Fu Q: USP1 inhibits NF-κB/NLRP3 induced pyroptosis through TRAF6 in osteoblastic MC3T3-E1 cells. J Musculoskelet Neuronal Interact. 22:536–545. 2022.PubMed/NCBI

61 

Williams SA, Maecker HL, French DM, Liu J, Gregg A, Silverstein LB, Cao TC, Carano RA and Dixit VM: USP1 deubiquitinates ID proteins to preserve a mesenchymal stem cell program in osteosarcoma. Cell. 146:918–930. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Chaugule S, Kim JM, Yang YS, Knobeloch KP, He X and Shim JH: Deubiquitinating enzyme USP8 is essential for skeletogenesis by regulating Wnt signaling. Int J Mol Sci. 22:102892021. View Article : Google Scholar : PubMed/NCBI

63 

Kaushal K, Tyagi A, Karapurkar JK, Kim EJ, Tanguturi P, Kim KS, Jung HS and Ramakrishna S: Genome-wide CRISPR/Cas9-Based screening for deubiquitinase subfamily identifies ubiquitin-specific protease 11 as a novel regulator of osteogenic differentiation. Int J Mol Sci. 23:8562022. View Article : Google Scholar : PubMed/NCBI

64 

Guo YC, Wang MY, Zhang SW, Wu YS, Zhou CC, Zheng RX, Shao B, Wang Y, Xie L, Liu WQ, et al: Ubiquitin-specific protease USP34 controls osteogenic differentiation and bone formation by regulating BMP2 signaling. EMBO J. 37:e993982018. View Article : Google Scholar : PubMed/NCBI

65 

Kim JY, Lee JM and Cho JY: Ubiquitin C-terminal hydrolase-L3 regulates Smad1 ubiquitination and osteoblast differentiation. FEBS Lett. 585:1121–1126. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Huang P, Yan R, Zhang X, Wang L, Ke X and Qu Y: Activating Wnt/β-catenin signaling pathway for disease therapy: Challenges and opportunities. Pharmacol Ther. 196:79–90. 2019. View Article : Google Scholar

67 

Zhou F, Li F, Fang P, Dai T, Yang B, van Dam H, Jia J, Zheng M and Zhang L: Ubiquitin-specific protease 4 antagonizes osteoblast differentiation through dishevelled. J Bone Miner Res. 31:1888–8198. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Guasto A and Cormier-Daire V: Signaling pathways in bone development and their related skeletal dysplasia. Int J Mol Sci. 22:43212021. View Article : Google Scholar : PubMed/NCBI

69 

Herhaus L and Sapkota GP: The emerging roles of deubiquitylating enzymes (DUBs) in the TGFβ and BMP pathways. Cell Signal. 26:2186–2192. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Baek D, Park KH, Lee KM, Jung S, Joung S, Kim J and Lee JW: Ubiquitin-specific protease 53 promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. Cell Death Dis. 12:2382021. View Article : Google Scholar : PubMed/NCBI

71 

Hariri H, Kose O, Bezdjian A, Daniel SJ and St-Arnaud R: USP53 regulates bone homeostasis by controlling rankl expression in osteoblasts and bone marrow adipocytes. J Bone Miner Res. 38:578–596. 2023. View Article : Google Scholar : PubMed/NCBI

72 

Tsuru M, Ono A, Umeyama H, Takeuchi M and Nagata K: Ubiquitin-dependent proteolysis of CXCL7 leads to posterior longitudinal ligament ossification. PLoS One. 13:e01962042018. View Article : Google Scholar : PubMed/NCBI

73 

Lin L, Li S, Hu S, Yu W, Jiang B, Mao C, Li G, Yang R, Miao X, Jin M, et al: UCHL1 impairs periodontal ligament stem cell osteogenesis in periodontitis. J Dent Res. 102:61–71. 2023. View Article : Google Scholar

74 

Cao Y, Zhang X, Hu M, Yang S, Li X, Han R, Zhou J, Li D and Liu D: CYLD inhibits osteoclastogenesis to ameliorate alveolar bone loss in mice with periodontitis. J Cell Physiol. 238:1036–1045. 2023. View Article : Google Scholar : PubMed/NCBI

75 

Ang E, Pavlos NJ, Rea SL, Qi M, Chai T, Walsh JP, Ratajczak T, Zheng MH and Xu J: Proteasome inhibitors impair RANKL-induced NF-kappaB activity in osteoclast-like cells via disruption of p62, TRAF6, CYLD, and IkappaBalpha signaling cascades. J Cell Physiol. 220:450–459. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Jin W, Chang M, Paul EM, Babu G, Lee AJ, Reiley W, Wright A, Zhang M, You J and Sun SC: Deubiquitinating enzyme CYLD negatively regulates RANK signaling and osteoclastogenesis in mice. J Clin Invest. 118:1858–1866. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Hong JY, Bae WJ, Yi JK, Kim GT and Kim EC: Anti-inflammatory and anti-osteoclastogenic effects of zinc finger protein A20 overexpression in human periodontal ligament cells. J Periodontal Res. 51:529–539. 2016. View Article : Google Scholar

78 

Lee EG, Boone DL, Chai S, Libby SL, Chien M, Lodolce JP and Ma A: Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 289:2350–2354. 2000. View Article : Google Scholar : PubMed/NCBI

79 

Lee MJ, Lim E, Mun S, Bae S, Murata K, Ivashkiv LB and Park-Min KH: Intravenous immunoglobulin (IVIG) attenuates TNF-induced pathologic bone resorption and suppresses osteoclastogenesis by inducing A20 expression. J Cell Physiol. 231:449–458. 2016. View Article : Google Scholar :

80 

Martens A and van Loo G: A20 at the crossroads of cell death, inflammation, and autoimmunity. Cold Spring Harb Perspect Biol. 12:a0364182020. View Article : Google Scholar

81 

Yan K, Wu C, Ye Y, Li L, Wang X, He W, Ren S and Xu Y: A20 inhibits osteoclastogenesis via TRAF6-dependent autophagy in human periodontal ligament cells under hypoxia. Cell Prolif. 53:e127782020. View Article : Google Scholar : PubMed/NCBI

82 

Birol M and Echalier A: Structure and function of MPN (Mpr1/Pad1 N-terminal) domain-containing proteins. Curr Protein Pept Sci. 15:504–517. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Fiore A, Liang Y, Lin YH, Tung J, Wang H, Langlais D and Nijnik A: Deubiquitinase MYSM1 in the hematopoietic system and beyond: A current review. Int J Mol Sci. 21:30072020. View Article : Google Scholar : PubMed/NCBI

84 

Li P, Yang YM, Sanchez S, Cui DC, Dang RJ, Wang XY, Lin QX, Wang Y, Wang C, Chen DF, et al: Deubiquitinase MYSM1 Is essential for normal bone formation and mesenchymal stem cell differentiation. Sci Rep. 6:222112016. View Article : Google Scholar : PubMed/NCBI

85 

Haffner-Luntzer M, Kovtun A, Fischer V, Prystaz K, Hainzl A, Kroeger CM, Krikki I, Brinker TJ, Ignatius A and Gatzka M: Loss of p53 compensates osteopenia in murine Mysm1 deficiency. FASEB J. 32:1957–1968. 2018. View Article : Google Scholar

86 

Lin YC, Zheng G, Liu HT, Wang P, Yuan WQ, Zhang YH, Peng XS, Li GJ, Wu YF and Shen HY: USP7 promotes the osteoclast differentiation of CD14+ human peripheral blood monocytes in osteoporosis via HMGB1 deubiquitination. J Orthop Translat. 40:80–91. 2023. View Article : Google Scholar : PubMed/NCBI

87 

Xie Z, Wu Y, Shen Y, Guo J, Yuan P, Ma Q, Wang S, Jie Z, Zhou H, Fan S and Chen S: USP7 inhibits osteoclastogenesis via dual effects of attenuating TRAF6/TAK1 axis and stimulating STING signaling. Aging Dis. 14:2267–2283. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Mustachio LM, Lu Y, Kawakami M, Roszik J, Freemantle SJ, Liu X and Dmitrovsky E: Evidence for the ISG15-specific deubiquitinase USP18 as an antineoplastic target. Cancer Res. 78:587–592. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Liu X, Lu Y, Chen Z, Liu X, Hu W, Zheng L, Chen Y, Kurie JM, Shi M, Mustachio LM, et al: The ubiquitin-specific peptidase USP18 promotes lipolysis, fatty acid oxidation, and lung cancer growth. Mol Cancer Res. 19:667–677. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Yim HY, Park C, Lee YD, Arimoto K, Jeon R, Baek SH, Zhang DE, Kim HH and Kim KI: Elevated response to type I IFN enhances RANKL-mediated osteoclastogenesis in Usp18-knockout mice. J Immunol. 196:3887–3895. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Li Q, Wang M, Xue H, Liu W, Guo Y, Xu R, Shao B and Yuan Q: Ubiquitin-specific protease 34 inhibits osteoclast differentiation by regulating NF-κB signaling. J Bone Miner Res. 35:1597–1608. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Li C, Qiu M, Chang L, Qi J, Zhang L, Ryffel B and Deng L: The osteoprotective role of USP26 in coordinating bone formation and resorption. Cell Death Differ. 29:1123–1136. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Zhu Q, Fu Y, Cui CP, Ding Y, Deng Z, Ning C, Hu F, Qiu C, Yu B, Zhou X, et al: OTUB1 promotes osteoblastic bone formation through stabilizing FGFR2. Signal Transduct Target Ther. 8:1422023. View Article : Google Scholar : PubMed/NCBI

94 

LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ and Siris ES: The clinician's guide to prevention and treatment of osteoporosis. Osteoporos Int. 33:2049–2102. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Eastell R, Rosen CJ, Black DM, Cheung AM, Murad MH and Shoback D: Pharmacological management of osteoporosis in postmenopausal women: An endocrine society* clinical practice guideline. J Clin Endocrinol Metab. 104:1595–1622. 2019. View Article : Google Scholar : PubMed/NCBI

96 

McClung M, Harris ST, Miller PD, Bauer DC, Davison KS, Dian L, Hanley DA, Kendler DL, Yuen CK and Lewiecki EM: Bisphosphonate therapy for osteoporosis: Benefits, risks, and drug holiday. Am J Med. 126:13–20. 2013. View Article : Google Scholar

97 

Shane E, Burr D, Abrahamsen B, Adler RA, Brown TD, Cheung AM, Cosman F, Curtis JR, Dell R, Dempster DW, et al: Atypical subtrochanteric and diaphyseal femoral fractures: Second report of a task force of the American society for bone and mineral research. J Bone Miner Res. 29:1–23. 2014. View Article : Google Scholar

98 

Deeks ED: Denosumab: A review in postmenopausal osteoporosis. Drugs Aging. 35:163–173. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Morin SN, Feldman S, Funnell L, Giangregorio L, Kim S, McDonald-Blumer H, Santesso N, Ridout R, Ward W, Ashe MC, et al: Clinical practice guideline for management of osteoporosis and fracture prevention in Canada: 2023 Update. CMAJ. 195:E1333–E1348. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Lobo RA, Pickar JH, Stevenson JC, Mack WJ and Hodis HN: Back to the future: Hormone replacement therapy as part of a prevention strategy for women at the onset of menopause. Atherosclerosis. 254:282–290. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Gosset A, Pouillès JM and Trémollieres F: Menopausal hormone therapy for the management of osteoporosis. Best Pract Res Clin Endocrinol Metab. 35:1015512021. View Article : Google Scholar : PubMed/NCBI

102 

Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, et al: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results from the women's health initiative randomized controlled trial. JAMA. 288:321–333. 2002. View Article : Google Scholar : PubMed/NCBI

103 

Prior JC, Seifert-Klauss VR, Giustini D, Adachi JD, Kalyan S and Goshtasebi A: Estrogen-progestin therapy causes a greater increase in spinal bone mineral density than estrogen therapy-a systematic review and meta-analysis of controlled trials with direct randomization. J Musculoskelet Neuronal Interact. 17:146–154. 2017.PubMed/NCBI

104 

de Villiers TJ, Hall JE, Pinkerton JV, Pérez SC, Rees M, Yang C and Pierroz DD: Revised global consensus statement on menopausal hormone therapy. Maturitas. 91:153–155. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Levin VA, Jiang X and Kagan R: Estrogen therapy for osteoporosis in the modern era. Osteoporos Int. 29:1049–1055. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Abdi F, Mobedi H, Bayat F, Mosaffa N, Dolatian M and Ramezani Tehrani F: The effects of transdermal estrogen delivery on bone mineral density in postmenopausal women: A meta-analysis. Iran J Pharm Res. 16:380–389. 2017.PubMed/NCBI

107 

Mosca L: Rationale and overview of the raloxifene use for the heart (RUTH) trial. Ann N Y Acad Sci. 949:181–185. 2001. View Article : Google Scholar

108 

Ensrud K, Genazzani AR, Geiger MJ, McNabb M, Dowsett SA, Cox DA and Barrett-Connor E: Effect of raloxifene on cardiovascular adverse events in postmenopausal women with osteoporosis. Am J Cardiol. 97:520–527. 2006. View Article : Google Scholar : PubMed/NCBI

109 

Vahle JL, Long GG, Sandusky G, Westmore M, Ma YL and Sato M: Bone neoplasms in F344 rats given teriparatide [rhPTH(1-34)] are dependent on duration of treatment and dose. Toxicol Pathol. 32:426–438. 2004. View Article : Google Scholar : PubMed/NCBI

110 

Andrews EB, Gilsenan AW, Midkiff K, Sherrill B, Wu Y, Mann BH and Masica D: The US postmarketing surveillance study of adult osteosarcoma and teriparatide: Study design and findings from the first 7 years. J Bone Miner Res. 27:2429–2437. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Xie J, Guo J, Kanwal Z, Wu M, Lv X, Ibrahim NA, Li P, Buabeid MA, Arafa EA and Sun Q: Calcitonin and bone physiology: In vitro, in vivo, and clinical investigations. Int J Endocrinol. 2020:32368282020. View Article : Google Scholar : PubMed/NCBI

112 

Holdsworth G, Roberts SJ and Ke HZ: Novel actions of sclerostin on bone. J Mol Endocrinol. 62:R167–R185. 2019. View Article : Google Scholar

113 

Weaver CM, Alexander DD, Boushey CJ, Dawson-Hughes B, Lappe JM, LeBoff MS, Liu S, Looker AC, Wallace TC and Wang DD: Calcium plus vitamin D supplementation and risk of fractures: An updated meta-analysis from the national osteoporosis foundation. Osteoporos Int. 27:367–376. 2016. View Article : Google Scholar :

114 

Meng J, Zhang W, Wang C, Zhang W, Zhou C, Jiang G, Hong J, Yan S and Yan W: Catalpol suppresses osteoclastogenesis and attenuates osteoclast-derived bone resorption by modulating PTEN activity. Biochem Pharmacol. 171:1137152020. View Article : Google Scholar

115 

Ferlazzo N, Andolina G, Cannata A, Costanzo MG, Rizzo V, Currò M, Ientile R and Caccamo D: Is melatonin the cornucopia of the 21st century? Antioxidants (Basel). 9:10882020. View Article : Google Scholar : PubMed/NCBI

116 

Lian C, Wu Z, Gao B, Peng Y, Liang A, Xu C, Liu L, Qiu X, Huang J, Zhou H, et al: Melatonin reversed tumor necrosis factor-alpha-inhibited osteogenesis of human mesenchymal stem cells by stabilizing SMAD1 protein. J Pineal Res. 61:317–327. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Zheng ZG, Cheng HM, Zhou YP, Zhu ST, Thu PM, Li HJ, Li P and Xu X: Dual targeting of SREBP2 and ERRα by carnosic acid suppresses RANKL-mediated osteoclastogenesis and prevents ovariectomy-induced bone loss. Cell Death Differ. 27:2048–2065. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Zheng HL, Xu WN, Zhou WS, Yang RZ, Chen PB, Liu T, Jiang LS and Jiang SD: Beraprost ameliorates postmenopausal osteoporosis by regulating Nedd4-induced Runx2 ubiquitination. Cell Death Dis. 12:4972021. View Article : Google Scholar : PubMed/NCBI

119 

Zhang Y, Wang C, Cao Y, Gu Y and Zhang L: Selective compounds enhance osteoblastic activity by targeting HECT domain of ubiquitin ligase Smurf1. Oncotarget. 8:50521–50533. 2016. View Article : Google Scholar : PubMed/NCBI

120 

Ye LC, Qian LF, Liang L, Jiang LJ, Che ZY and Guo YH: Overexpression of miR-195-5p reduces osteoporosis through activating BMP-2/SMAD/Akt/RUNX2 pathway via targeting SMURF1. J Biol Regul Homeost Agents. 35:1201–1216. 2021.

121 

Oyajobi BO, Garrett IR, Gupta A, Flores A, Esparza J, Muñoz S, Zhao M and Mundy GR: Stimulation of new bone formation by the proteasome inhibitor, bortezomib: implications for myeloma bone disease. Br J Haematol. 139:434–438. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Uyama M, Sato MM, Kawanami M and Tamura M: Regulation of osteoblastic differentiation by the proteasome inhibitor bortezomib. Genes Cells. 17:548–558. 2012. View Article : Google Scholar : PubMed/NCBI

123 

Fang Y, Liu Y, Zhao Z, Lu Y, Shen X, Zhu T, Hou M, He F, Yang H, Zhang Y, et al: Bortezomib rescues ovariectomy-induced bone loss via SMURF-mediated ubiquitination pathway. Oxid Med Cell Longev. 2021:96612002021. View Article : Google Scholar

124 

Khedgikar V, Kushwaha P, Gautam J, Verma A, Changkija B, Kumar A, Sharma S, Nagar GK, Singh D, Trivedi PK, et al: Withaferin A: A proteasomal inhibitor promotes healing after injury and exerts anabolic effect on osteoporotic bone. Cell Death Dis. 4:e7782013. View Article : Google Scholar : PubMed/NCBI

125 

Kabekkodu SP, Shukla V, Varghese VK, D' Souza J, Chakrabarty S and Satyamoorthy K: Clustered miRNAs and their role in biological functions and diseases. Biol Rev Camb Philos Soc. 93:1955–1986. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Huang Y, Yang Y, Wang J, Yao S, Yao T, Xu Y, Chen Z, Yuan P, Gao J, Shen S and Ma J: miR-21-5p targets SKP2 to reduce osteoclastogenesis in a mouse model of osteoporosis. J Biol Chem. 296:1006172021. View Article : Google Scholar : PubMed/NCBI

127 

Liu C, Gao X, Li Y, Sun W, Xu Y, Tan Y, Du R, Zhong G, Zhao D, Liu Z, et al: The mechanosensitive lncRNA Neat1 promotes osteoblast function through paraspeckle-dependent Smurf1 mRNA retention. Bone Res. 10:182022. View Article : Google Scholar : PubMed/NCBI

128 

Jiang Y, Wu W, Jiao G, Chen Y and Liu H: LncRNA SNHG1 modulates p38 MAPK pathway through Nedd4 and thus inhibits osteogenic differentiation of bone marrow mesenchymal stem cells. Life Sci. 228:208–214. 2019. View Article : Google Scholar : PubMed/NCBI

129 

Liu L, Jin R, Duan J, Yang L, Cai Z, Zhu W, Nie Y, He J, Xia C, Gong Q, et al: Bioactive iron oxide nanoparticles suppress osteoclastogenesis and ovariectomy-induced bone loss through regulating the TRAF6-p62-CYLD signaling complex. Acta Biomater. 103:281–292. 2020. View Article : Google Scholar

130 

Liu Y, Huang X, He X, Zhou Y, Jiang X, Chen-Kiang S, Jaffrey SR and Xu G: A novel effect of thalidomide and its analogs: Suppression of cereblon ubiquitination enhances ubiquitin ligase function. FASEB J. 29:4829–4839. 2015. View Article : Google Scholar : PubMed/NCBI

131 

Mishima K, Kitoh H, Ohkawara B, Okuno T, Ito M, Masuda A, Ishiguro N and Ohno K: Lansoprazole upregulates polyubiquitination of the TNF receptor-associated factor 6 and facilitates Runx2-mediated osteoblastogenesis. EBioMedicine. 2:2046–2061. 2015. View Article : Google Scholar

132 

Li X, Sun W, Li J, Wang M, Zhang H, Pei L, Boyce BF, Wang Z and Xing L: Clomipramine causes osteoporosis by promoting osteoclastogenesis via E3 ligase Itch, which is prevented by zoledronic acid. Sci Rep. 7:413582017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 54 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan X, Zhang R, Xu G, Fan P, Luo W, Cai C and Ge R: Role of ubiquitination in the occurrence and development of osteoporosis (Review). Int J Mol Med 54: 68, 2024
APA
Fan, X., Zhang, R., Xu, G., Fan, P., Luo, W., Cai, C., & Ge, R. (2024). Role of ubiquitination in the occurrence and development of osteoporosis (Review). International Journal of Molecular Medicine, 54, 68. https://doi.org/10.3892/ijmm.2024.5392
MLA
Fan, X., Zhang, R., Xu, G., Fan, P., Luo, W., Cai, C., Ge, R."Role of ubiquitination in the occurrence and development of osteoporosis (Review)". International Journal of Molecular Medicine 54.2 (2024): 68.
Chicago
Fan, X., Zhang, R., Xu, G., Fan, P., Luo, W., Cai, C., Ge, R."Role of ubiquitination in the occurrence and development of osteoporosis (Review)". International Journal of Molecular Medicine 54, no. 2 (2024): 68. https://doi.org/10.3892/ijmm.2024.5392