Open Access

Otubain 2 stabilizes TNF‑receptor associated factor 3 to accelerate bone fracture healing

  • Authors:
    • Li Zhang
    • Jialiang Guo
    • Shan Feng
    • Yue Zheng
    • Haixu Wang
    • Huijie Ma
    • Wei Chen
    • Yingze Zhang
    • Zhiyong Hou
  • View Affiliations

  • Published online on: June 5, 2025     https://doi.org/10.3892/mmr.2025.13588
  • Article Number: 223
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

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


Abstract

Increased incidence of traumatic fracture markedly effects the quality of life of patients. Using a rat model of femur fracture, the present study aimed to investigate the effects of otubain 2 (OTUB2), a deubiquitinating enzyme, on bone fracture healing. Bone marrow mesenchymal stem cells (BMSCs) were harvested from the marrow cavity of rat femurs and tibiae and subsequently subjected to osteogenic differentiation in vitro. Results of the present study revealed that lentivirus‑mediated OTUB2 overexpression accelerated rat bone fracture healing, potentiated fracture callus formation and cartilaginous ossification and regulated the expression of proteins associated with bone remodeling. In addition, OTUB2 overexpression facilitated the osteogenic differentiation and mineralization of BMSCs and promoted the expression of TNF‑receptor associated factor 3 (TRAF3) both in vivo and in vitro. Co‑immunoprecipitation analysis was used to verify the physical interaction between OTUB2 and TRAF3 and further results demonstrated that OTUB2 reduced the ubiquitination of TRAF3. The results of the present study also demonstrated that TRAF3 knockdown repressed the OTUB2‑induced osteogenic differentiation and mineralization of BMSCs. Collectively, these results demonstrated that OTUB2 may stabilize TRAF3 to accelerate bone fracture healing.

Introduction

Bone is one of the few tissues that heals without forming a fibrous scar (1). The term ‘bone fracture’ is used to describe the destruction of bone continuity or breakdown of the bone structural integrity (2). Bone heals over a period of time (1); however, open fractures are vulnerable to complications, such as malunion, delayed union, non-union and refracture (3,4). In total, ~5–10% of fractures will show delayed healing (5). At present, the incidence of traumatic fracture continues to increase, current estimates for traumatic fractures are 23.3% for men and 11.2% for women (6), and this markedly affects the quality of life of patients (7,8). Fracture healing is a dynamic process. The initial stage includes the hematoma, which generates an inflammatory environment. Next, endochondral ossification, removal and calcification of the endochondral cartilage indicate the fracture healing has reached the middle to late stage (9). Then, the chronic remodeling concludes the healing process (9).

The ubiquitin-proteasome system is a major pathway of protein post-translational modification, mediated by E1, E2 and E3 ubiquitin ligases, in addition to deubiquitinating enzymes (10). Ubiquitination is a reversible process; whereby deubiquitinating enzymes remove ubiquitin from poly-ubiquitin chains and targeted proteins (11). Otubain (OTUB)2 is a member of the OTUB superfamily of deubiquitinylases that inhibits the ubiquitination of substrates and enhance protein stability (12,13). Dysregulation of the OTUB protein family may lead to bone dysplasia (13). Stanišić et al (14) reported that OTUB1 weakens the stability of estrogen receptor α (14), which serves an essential role in skeletal formation (15). Thus, OTUB proteins may act as a vital part in the pathophysiology of bone development and homeostasis, which refers to the balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption (16). Results of the previous study revealed that osteoblast activity and osteogenesis were enhanced during fracture healing. In addition, osteoclast function and bone resorption were reduced, promoting the formation of an external callus used to stabilize bone fragments (17). Li et al (18) reported that OTUB2 knockdown reverses Shh- or Smo-induced upregulation of runt related transcription factor 2 (RUNX2), bone morphogenetic protein 2 and tissue nonspecific alkaline phosphatase (TNAP), key regulators of bone formation (18). Thus, OTUB2 may exhibit potential in osteogenesis and bone fracture healing (18). Results of a previous study demonstrate that fracture healing may also be mediated by the ubiquitination of genes associated with osteogenesis (19).

The present study aimed to investigate the effects of OTUB2 on bone fracture healing both in vivo and in vitro. A model of open fracture was established in the femora of Sprague Dawley rats, as previously described (4). Notably, bone callus formation requires the osteoblastic differentiation of bone marrow mesenchymal stem cells (BMSCs) (20,21) and previous studies demonstrate that this differentiation promotes bone fracture healing (22,23). Thus, BMSCs were obtained from the rat marrow cavity of femurs and tibiae and subsequently subjected to osteogenic differentiation in vitro (24). The functions of OTUB2 in bone fracture healing, histological damage, bone formation and mineralization were further explored.

Materials and methods

Animal experiments

Animal experiments were approved by the Laboratory Animal Ethical and Welfare Committee of Hebei Medical University (Hebei, China; approval no. IACUC-Hebmu-2021007), following The Guideline for the Care and Use of Laboratory Animals (25).

Bioinformatics analysis

Proteins that interact with OTUB2 were analyzed using the STRING database (https://cn.string-db.org/). Gene Ontology (GO) enrichment analysis of proteins that interacted with OTUB2 was carried out using the DAVID database (https://david.ncifcrf.gov/home.jsp).

Lentivirus preparation and infection

A 2nd generation lentiviral vector system was used. For OTUB2 overexpression, OTUB2 cDNA was amplified and subcloned into the lentivirus vector (LV), pLVX–IRES-Puro (Unibio) and referred to as LV-OTUB2. pLVX–IRES-Puro with no cDNA insertion was used as the negative control (NC) for LV-OTUB2 and referred to as LV-NC. For TNF-receptor associated factor 3 (TRAF3) knockdown, TRAF3 short hairpin RNA (shRNA) was synthetized and subcloned into pLVX-shRNA1 (Unibio). shNC, a non-targeting shRNA, was subcloned into pLVX-shRNA1 and used as the NC for LV-shTRAF3. Subsequently, pLVX-OTUB2/pLVX-shTRAF3/NC, pSPAX2 and pMD2.G vectors were mixed at a ratio of 4:3:1 (14.0:10.5:3.5 µg in a 10 cm culture flask) and transfected into 293T cells (iCell Bioscience). Following transfection, recombinant virus-containing supernatants were collected and filtered using a 0.45-µm membrane. In the logarithmic phase of growth (multiplicity of infection, 20) BMSCs were infected with LV-NC, LV-OTUB2, LV-shNC or LV-shTRAF3, as previously described (26). Sequences of shTRAF3 and shNC were as follows: shTRAF3, 5′-ccgCGAAGACAGTGGAGGACAAGTttcaagagaACTTGTCCTCCACTGTCTTCGttttt-3′; and shNC, 5′-ccgTTCTCCGAACGTGTCACGTttcaagagaACGTGACACGTTCGGAGAAttttt-3′. Cells were further subjected to the osteogenic differentiation at 72 h post-transfection.

Fracture model

Male Sprague Dawley rats (age, 12 weeks; weight, 410±10 g) were obtained from Liaoning Changsheng Biotechnology. Rats were housed with food and water ad libitum in a humidity (50±10%) and temperature-controlled (22±1°C) environment for a week of acclimation under a 12/12 h light/dark cycle. The fracture model was established as previously described (4), Briefly, animals were subjected to inhalation of isoflurane for anesthesia, using an induction rate of 5% and a maintenance rate of 2%, as previously described (27). An incision was made on the middle lateral thigh of rats, followed by the blunt dissection of muscle around the femur. The femur was cut and a sterile Kirschner wire was inserted to stabilize the fracture. Subsequently, rats were randomly divided into three groups: i) Control; ii) LV-NC; and iii) LV-OTUB2. Rats in the LV-NC and LV-OTUB2 groups were injected with 1×108 transducing units/ml (50 µl/injection) of LV-NC or LV-OTUB2 at the fracture site and this was repeated once a week for 2, 4 or 8 weeks. Fractures of rats in the control group were left untreated. Rats were sacrificed with 70% vol/min CO2 at each time point unless they met a prior humane endpoint. All animals were evaluated, weighted and scored daily with set criteria as previously described (28,29). Briefly, any finding listed as a humane endpoint, a score of 3 in two or more categories and a total score >9 led to the animal being euthanised. The categories including general appearance, activity, hydration, respiration, ambulation, surgical complications and weight loss. Following euthanasia, cardiac and respiratory arrest, as well as fixed and dilated pupils were observed in the rats for ~10 min before the animals were confirmed as deceased. A total of 162 rats were used in the present study. Among them, 36 rats [18 rats for western blotting; 18 rats for X-radiography and reverse transcription-quantitative PCR (RT-qPCR); n=6 rats/group] were used at 2 weeks post fracture. A total of 54 rats (18 rats for western blotting; 18 rats for X-radiography and RT-qPCR; 18 rats for pathological staining; n=6 rats/group) were used at 4 weeks post fracture. A total of 72 rats (18 rats for western blotting; 18 rats for X-radiography and RT-qPCR; 18 rats for micro-CT and pathological staining; 18 rats for TNAP activity detection; n=6 rats/group) were used at 8 weeks post fracture.

Reverse transcription-quantitative (RT-q) PCR

Total RNA was extracted from the callus tissue of the femur of rats using TRIpure lysis solution (BioTeke Corporation) according to the manufacturer's protocols. Subsequently, total RNA was reverse transcribed into cDNA using the BeyoRT II kit (Beyotime Institute of Biotechnology) according to the manufacturer's instructions. Briefly, total RNA, oligo (dT), reaction buffer, RNase inhibitor, dNTP Mix and BeyoRT II M-MLV reverse transcriptase were used to form a reaction system. According to the manufacturers' instructions, the system was incubated at 42°C for 50 min, followed by incubation at 80°C for 10 min. Subsequently, cDNA was amplified using 2X Taq PCR MasterMix and SYBR Green PCR Master Mix (Beijing Solarbio Science & Technology Co., Ltd.) in accordance with manufacturer's instructions. The reaction system included 1 µl cDNA, 1 µl primer, 0.3 µl SYBR Green, 10 µl 2X Taq PCR MasterMix and 7.7 µl ddH2O. The thermocycling conditions used were as follows: 94°C for 5 min, followed by 40 cycles of 94°C for 15 sec, 60°C for 25 sec and 72°C for 30 sec. The relative expression of targeted genes was quantified using six independent measurements and the 2−ΔΔCq method (30). β-actin was used as the internal reference gene. Primer sequences are listed in Table I. The experiments were repeated thrice.

Table I.

Sequences of primers used in reverse transcription-quantitative PCR.

Table I.

Sequences of primers used in reverse transcription-quantitative PCR.

GeneSequence (5′-3′)
Otubain 2F: TCAATCCGAAAGACCAAA
R: TGTGAGGAGGCGTAAGAA
Tissue nonspecific alkaline phosphataseF: AGTCCGTGGGCATCGTG
R: CCTCTGGCGGCATCTCA
Runt related transcription factor 2F: CCATAACGGTCTTCACAAATC
R: GAGGCGGTCAGAGAACAAACT
OsteoprotegerinF: TCCCTTGCCCTGACTAC
R: CCTGAGAAGAACCCATCC
Receptor activator of nuclear factor-κB ligandF: CATCGGGTTCCCATAAAG
R: GAAGCAAATGTTGGCGTA
β-actinF: TGGCACCACACTTTCTACAATGAGC
R: GGGTCATCTTTTCACGGTTGG

[i] F, forward; R, reverse.

Western blotting analysis

Total protein was extracted from BMSCs or the callus tissue in the femur of rats using Radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology) mixed with phenylmethanesulfonyl fluoride (100:1; Beyotime Institute of Biotechnology). Samples were centrifuged at 10,000 × g for 5 min at 4°C and supernatants were collected. Total protein was quantified using a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. A total of 20 µg of protein was separated by SDS-PAGE s with a 5% stacking gel and a 10% running gel. Separated proteins were transferred onto PVDF membranes and blocked with 5% skimmed milk diluted in tris-buffered saline tween-20 for 1 h at room temperature. Following blocking, membranes were incubated with the following primary antibodies at 4°C overnight: Polyclonal rabbit anti-OTUB2 (1:500; Sabbiotech), polyclonal rabbit anti-TNAP (1:1,000; ABclonal Biotech, Co., Ltd.), polyclonal rabbit anti-RUNX2 (1:500; Proteintech Group, Inc.), polyclonal rabbit anti-osteoprotegerin (OPG; 1:1,000; Affinity Biosciences, Ltd.), polyclonal rabbit anti-receptor activator of nuclear factor-κB ligand (RANKL; 1:500; Affinity Biosciences, Ltd.), polyclonal rabbit anti-TRAF3 (1:1,000; ABclonal Biotech, Co., Ltd.), polyclonal rabbit anti-Flag (ABclonal Biotech, Co., Ltd.), polyclonal rabbit anti-ubiquitin (Proteintech Group, Inc.) and monoclonal mouse anti-β-actin (1:1,000; Santa Cruz Biotechnology, Inc.). Following primary incubation, membranes were incubated with the following secondary antibodies at 37°C for 45 min: Goat anti-rabbit horseradish peroxidase-conjugated IgG (1:5,000; Beyotime Institute of Biotechnology) and goat anti-mouse horseradish peroxidase-conjugated IgG (1:5,000; Beyotime Institute of Biotechnology). Protein bands were visualized using ECL reagent (Beyotime Institute of Biotechnology) on the WD-9413B gel imaging system (Liuyi Biotechnology). Six independent measurements were obtained using callus tissue samples and three independent measurements were obtained using BMSC samples. The band densities were quantified with the Gel-Pro-Analyzer software (version 4.0; Media Cybernetics, Inc.).

X-radiography and micro-computed tomography (CT) analysis

Fractured femurs were imaged using the CSM-2R X-ray apparatus (Softex) at 2-, 4- and 8-weeks post-fracture. Radiographic fracture healing was scored from Grade 1–6, as previously described (3). Briefly, fracture healing was scored as follows: i) Grade 1, no calcification; ii) Grade 2, patchy calcification; iii) Grade 3, calcification takes on the appearance of a callus; iv) Grade 4, callus bridging across the fracture gap; v) Grade 5, continuity of bone trabeculae; and vi) Grade 6, remodeling to normal bone.

Fractured femurs were scanned using the Quantum GX micro-CT imaging system (PerkinElmer, Inc.). Briefly, fractured femurs were scanned at 90 kV and 88 µA using a 50-µm voxel. Using the fracture line as the center, 50 slices were measured between the distal and proximal edges. The callus was segmented from the background as a region of interest. Subsequently, bone volume fraction (BV/TV) and bone mineral density (BMD) were auto-obtained using micro-CT analysis and 3D images were captured (31,32).

Histological analysis

Tissues were fixed in 10% formalin solution at 4°C for 48 h. The fixed tissues were rinsed in running water for 4 h at room temperature. Samples were dehydrated in gradient ethanol (50–100 %), embedded in paraffin and then cut into 5 µm sections using a microtome (cat. no. RM 2235; Leica Microsystems, Inc.). Paraffin-embedded sections of fracture callus were deparaffinized in xylene and rehydrated in an ethanol gradient with distilled water. Formation of callus was examined using H&E staining (33). Cartilaginous ossification was analyzed using safranine O-fast green staining. Briefly, sections were stained with safranine O for 5 min at room temperature, followed by rinsing with ethanol gradient. Sections were subsequently stained with fast green for 1 min at room temperature and imaged using a BX53 microscope in bright-field mode (Olympus Corporation).

Isolation and osteogenic differentiation of BMSCs

Isolation and osteogenic differentiation of BMSCs were performed as previously described (24). Briefly, femurs and tibiae were collected from rats and muscles surrounding the bone were removed. The marrow cavity was flushed with DMEM (Wuhan Servicebio Technology Co., Ltd.) for cell harvesting and cells were centrifuged at 300 × g for 7 min at room temperature. Subsequently, cells were cultured in DMEM supplemented with 10% fetal bovine serum (Sijiqing Biological Engineering Materials Co., Ltd.) at 37°C in an incubator with 5% CO2. BMSCs were cultured until the third passage was reached and used in subsequent experiments after ~2 weeks. BMSC phenotype was confirmed via flow cytometry with surface antigens.

Osteogenic differentiation was performed at 72 h post-transfection, as previously described (34). Briefly, cell medium was removed and replaced with medium for inducing osteogenic differentiation [100 nM dexamethasone (MilliporeSigma), 50 µM vitamin C (Sinopharm Chemical Reagent Co., Ltd.) and 10 mM β-sodium glycerophosphate (MilliporeSigma)]. Differentiation medium was changed every 2 days. At 3 days post-induction, total protein was extracted for western blotting analysis (35). At 7 days post-induction, TNAP activity was detected and at 14 days post-induction, Alizarin Red S staining was performed to assess calcium deposition, as previously described (35,36).

Flow cytometry

Following centrifugation at 300 × g for 7 min at 4°C, BMSCs were rinsed and suspended with 100 µl phosphate buffer saline. In total, 1×106 cells were incubated with 0.25 µg fluorescein isothiocyanate (FITC)-conjugated anti-CD45 (MultiSciences Biotech), 1 µg FITC-conjugated anti-CD29 (BioLegend, Inc.), 0.06 µg FITC-conjugated anti-CD90 (BioLegend, Inc.) and 0.25 µg PE-Cyanine 7-conjugated anti-CD31 (Thermo Scientific, Inc.) at 4°C in the dark. Subsequently, cells were analyzed via NovoCyte flow cytometry (ACEA Biosciences), using three independent measurements. Data was analyzed using NovoExpress software (version 1.4.1; Agilent Technologies, Inc.).

Alizarin Red S staining

Cells were fixed with 4% paraformaldehyde at room temperature for 15 min and stained with Alizarin Red for 30 min at room temperature. Images were captured using a microscope (IX53l; Olympus Corporation). Calcium deposits were quantified following the addition of 10% cetylpyridinium chloride (Shanghai Macklin Biochemical Co., Ltd.) for 15 min at room temperature. Absorbance was measured at a wavelength of 570 nm (37).

Detection of TNAP

The levels of TNAP were assessed using the TNAP Detection Kit (Wanleibio Co., Ltd.) according to the manufacturer's instructions. Briefly, ultrasonic cell disruption (300 W; ultrasonication for 3 sec with an interval of 30 sec repeated 5 times) was carried out in an ice bath and cells were subsequently centrifuged at 421 × g for 10 min at 4°C. In total, three independent supernatant samples were used for the detection of TNAP levels.

Callus tissues were homogenized with normal saline (1:9; g/v). The tissue homogenate was centrifuged at 421 × g at room temperature for 10 min. The resulting supernatant was obtained for the detection of TNAP levels. A total of six independent supernatant samples were used for the detection of TNAP levels.

Co-immunoprecipitation

Wild-type fragments of OTUB2 cDNA, wild-type TRAF3 cDNA and OTUB2 fragments with a point mutation at C51S were synthetized by General Biotech (Anhui) Co., Ltd. Subsequently, wild-type OTUB2 fragments and OTUB2 fragments with a point mutation were inserted into pcDNA3.1(+)_myc-His A vectors (General Biotech (Anhui) Co., Ltd.). Wild-type fragments of TRAF3 were inserted into p3×FLAG-CMV-10 vectors (General Biotech (Anhui) Co., Ltd.). All constructs were verified using enzyme digestion and sequencing. Subsequently, Lipofectamine® 3000 was used to transiently transfect OTUB2-Myc/OTUB2-Myc (C51S) and TRAF3-Flag plasmids into 293T cells. One day prior to transfection, cells were cultured in 6 well-plates until 80% confluence was reached. In addition, cells were starved for 2 h prior to transfection and subsequently incubated with OptiMEM containing 5 µl Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.), 5 µl P3000 reagent and 2 µg plasmids. Following 48 h of transfection, 293T cells were lysed with radioimmunoprecipitation assay buffer mixed with phenylmethanesulfonyl fluoride (100:1), followed by mixing in liquid nitrogen. Cells were centrifuged at 10,000 × g for 5 min at 4°C and the supernatant was collected and subjected to immunoprecipitation using a co-immunoprecipitation kit (cat. no. 26149; Thermo Fisher Scientific, Inc.). Briefly, 20 µl of resin slurry was used to pre-clear the lysates, which were further incubated with 1 µg of immobilized antibodies (Flag-tag or Myc-tag). Following elution and centrifugation (1,000 × g for 5 min at 4°C) to obtain the precipitates, proteins were used for subsequent western blot analysis.

Molecular docking analysis

OTUB2 and TRAF3 sequences were obtained from the National Center for Biotechnology Information (https://www.ncbi.nlm.nih.gov/) and crystal structures were obtained via homology modeling on the SWISS-MODEL database (https://swissmodel.expasy.org/). Interactions between proteins were forecasted using the GRAMM database (https://gramm.compbio.ku.edu/request) and a graphic representation of the protein-protein interaction was established using PyMOL software (version 2.0; Schrodinger). Grey was indicative of the OTUB2 protein, purple was indicative of the TRAF3 protein and stick structures of red and blue were indicative of binding. Notably, the yellow dotted line was indicative of a hydrogen bond.

Statistical analysis

Data are presented as the mean ± standard deviation, or as box and whisker plots, with the ‘box’ depicting the median, 1st quartile and 3rd quartile and the ‘whisker’ depicting the standard deviation. Data were analyzed using GraphPad Prism software (version, 8.0; GraphPad; Dotmatics). In vitro experiments were independently repeated three times and in vivo experiments were independently repeated six times (6 rats/group). Differences between multiple groups were analyzed using one-way ANOVA followed by Tukey's post hoc test and comparisons between two groups were analyzed using an unpaired Student's t-test. Effect size was calculated to determine the power of the analysis, due to the small sample size included in the present study, as previously described (38). The sample size of animals was chosen according to the results of power analysis. The priori/post-hoc power analysis was carried out using G*power 3.1.9.7 software (Franz Faul). It was found appropriate to complete the study with at least 6 samples for each group (α err probe was 0.05; effect size was 0.9; power was 0.8). Previous studies reported that Student's t-tests are suitable when sample size is small and effect size is large (39,40). In addition, bone fracture healing scores were analyzed using a Kruskal-Wallis test followed by Dunn's post hoc analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

OTUB2 accelerates bone fracture healing

To investigate the potential effects of OTUB2 on bone fracture healing, a fracture model was established through the injection of lentivirus targeting OTUB2 around the fracture sites of rats (Fig. 1A). At 2-, 4- and 8-weeks post-fracture, mRNA and protein expression levels of OTUB2 were elevated in the fracture callus of rats in the LV-OTUB2 group (Fig. 1B and C). The effects of OTUB2 overexpression were subsequently investigated using X-radiography and micro-CT analysis. Results of the present study revealed that the speed of fracture healing was increased and fracture healing remodeling was improved in the LV-OTUB2 group at 8 weeks post-fracture, when compared with the LV-NC group (Fig. 1D and E). In addition, the elevated callus area was observed at the fracture sites (Fig. 1D). Results of the present study also revealed that specimens exhibited severe loss and erosion of the femur following bone fracture; however, this was alleviated following OTUB2 overexpression. Representative micro-CT 3D images of fractured femur are displayed in Fig. 1F. In addition, quantification of the micro-CT 3D analysis demonstrated that BMD and BV/TV were increased following OTUB2 overexpression, indicative of accelerated bone fracture healing and bone formation (Fig. 1G). These results highlighted that OTUB2 may promote bone fracture healing.

OTUB2 regulates bone callus formation, cartilaginous ossification and bone remodeling

The potential effects of OTUB2 on bone callus formation and cartilaginous ossification were further investigated in the present study. Results of the H&E staining analysis demonstrated that OTUB2 overexpression was associated with improved recovery at the fracture sites, with the formation of woven bone at 4- and 8-weeks post-fracture, compared with the LV-NC group (Fig. 2A). In addition, OTUB2 overexpression promoted soft callus formation at 4- and 8-weeks post-fracture. OTUB2 overexpression was associated with the domination of woven bone at the fracture sites at 8 weeks post-fracture and this was close to union (Fig. 2A). Results of the safranine O-fast green staining analysis revealed that the cartilage area of rats in the OTUB2 overexpression group was reduced at 4- and 8-weeks post-fracture, compared with the LV-NC group (Fig. 2B). At week 8, a few calcified cartilages and a limited number of uncalcified cartilages were observed in the fracture calluses of rats in the OTUB2 overexpression group (Fig. 2B), with ossification of the cartilage callus. Collectively, these data suggested that OTUB2 may potentiate fracture callus formation and cartilaginous ossification.

Bone formation and mineralization, cartilage maturation and bone mass play vital roles in the bone remodeling of fracture. Thus, genes and proteins associated with bone fracture were investigated at 2 weeks post-fracture. Results of the present study revealed that OTUB2 overexpression was associated with increased TNAP activity (Fig. 2C). In addition, OTUB2 overexpression was associated with increased TNAP and RUNX2 mRNA expression levels and an increase in the ratio between OPG and RANKL expression (Fig. 2D-F). Notably, results of the western blot analysis were comparable with those obtained using RT-qPCR (Fig. 2G and H).

OTUB2 facilitates the osteogenic differentiation and mineralization of BMSCs

Results of the present study revealed that OTUB2 exhibited protective effects on bone fracture healing in vivo. Thus, it was hypothesized that OTUB2 may play a similar role in vitro. In the present study, a model of osteogenic differentiation was established using BMSCs (Fig. 3A). BMSCs were isolated from rats and the phenotype was identified using flow cytometry with surface antigens. Results of the present study exhibited positivity for antigens CD29 and CD90 and negativity for antigens CD45 and CD31, indicative of a high cell purity (Fig. 3B). Notably, OTUB2 expression was increased in BMSCs infected with LV-OTUB2 (Fig. 3C). Results of the Alizarin Red S staining analysis revealed that OTUB2 potentiated the mineralization of BMSCs, with an increased number of calcified nodules that were bright red in color (Fig. 3D and E). In addition, OTUB2 overexpression was associated with increased TNAP expression levels (Fig. 3F), increased RUNX2 and OPG expression levels and reduced RANKL expression levels (Fig. 3G and H). Collectively, these results suggested that OTUB2 may facilitate the osteogenic differentiation and mineralization of BMSCs.

OTUB2 deubiquitinates the TRAF3 protein

The present study aimed to determine the specific mechanisms underlying the OTUB2-induced increases in osteogenesis and mineralization. Notably, deubiquitinase OTUB2 interacts with substrate proteins to remove covalently-attached ubiquitin, thereby controlling substrate abundance. Thus, it was hypothesized that the protective role of OTUB2 may be mediated by the stability of downstream substrates and proteins that interacted with OTUB2 were analyzed using the STRING database. As displayed in Fig. 4A, results of the present study revealed 10 proteins that interacted with OTUB2. To further investigate the specific functions of these proteins, GO enrichment analysis was performed (Fig. 4B). Results of the present study revealed that the OTUB2-interacting proteins were enriched in ‘protein deubiquitination’, ‘ubiquitin-dependent protein catabolic process’ and ‘protein modification by small protein conjugated or removal’. These enriched terms provided direction for further investigation of the mechanisms downstream of OTUB2 and TRAF3, a protein enriched in ‘regulation of protein polyubiquitination’ was selected for subsequent analyses (Fig. 4C). As a member of the TRAF family, TRAF3 plays a role in promoting bone formation and remodeling (41). Thus, it was hypothesized that the protective effects of OTUB2 in fracture healing and the osteogenic differentiation of BMSCs may be associated with TRAF3. In the present study, co-immunoprecipitation analysis was used to verify the physical interaction between OTUB2 and TRAF3 in 293T cells (Fig. 4D and E). Results of the protein molecular docking analysis revealed a hydrogen bond between OTUB2 and the TRAF3 protein, indicative of binding (Fig. 4F). In addition, results of the present study revealed that OTUB2 reduced the ubiquitination of TRAF3 (Fig. 4G). Subsequently, HEK293 cells expressing inactive enzyme mutant OTUB2 C51S were generated and results of the present study revealed that the OTUB2 mutation enhanced the ubiquitination of TRAF3 (Fig. 4H). These results highlighted that OTUB2 may repress TRAF3 ubiquitination and this is dependent on the deubiquitinase activity.

TRAF3 knockdown represses the osteogenic differentiation and mineralization of BMSCs

As displayed in Fig. 2G and 3G, TRAF3 protein expression levels were increased following OTUB2 overexpression in vivo and in vitro. As a downstream factor of OTUB2, the role of TRAF3 attracted our interest. It was found that TNAP activity was reduced following TRAF3 knockdown (Fig. 5A) and the mineralization of BMSCs was markedly repressed (Fig. 5B and C). Results of the present study also revealed that TRAF3 knockdown reduced the expression levels of RUNX2 and OPG and promoted RANKL expression (Fig. 5D and E). Collectively, these results demonstrated that TRAF3 knockdown may reduce the osteogenic differentiation of BMSCs.

OTUB2 promotes the osteogenic differentiation and mineralization of BMSCs through increased TRAF3 expression

In the present study, TRAF3 expression was reduced following transfection with LV-shTRAF3 (Fig. 6A). Results of the present study revealed that TNAP expression levels were reduced and the mineralization of BMSCs with OTUB2 overexpression was repressed following TRAF3 downregulation (Fig. 6B-D). Collectively, these results demonstrated that OTUB2 may promote the osteogenic differentiation and mineralization of BMSCs through upregulation of TRAF3 (Fig. 6E).

Discussion

At present, the increasing incidence of traumatic fracture markedly affects the quality of life of patients. Notably, elderly patients who have experienced a fracture exhibit other comorbidities, including pulmonary embolism, infection and heart failure, which may lead to an increased risk of mortality (42). Results of the present study highlighted that further investigations are required to determine the specific mechanisms underlying fracture. To the to the best of the authors' knowledge, the present study was the first to demonstrate the protective effects of OTUB2 in bone fracture healing. Notably, OTUB2 may serve a role in facilitating bone callus formation, cartilaginous ossification and bone remodeling, both in vitro and in vivo. Results of the present study revealed that OTUB2 facilitated the osteogenic differentiation and mineralization of BMSCs and this was mediated by the deubiquitination of the TRAF3 protein.

Bone formation and mineralization, cartilage maturation and bone mass serve a vital role in bone remodeling of fracture, thus, genes and proteins associated with bone fracture were investigated. Notably, OPG and RANKL are expressed by osteoblasts and RANKL regulates osteoclastogenesis through binding to RANK secreted by osteoclasts (43). On the other hand, OPG is a decoy receptor of RANKL that protects cells from osteoclast formation (43). Thus, the ratio between OPG/RANKL is crucial in determining bone mass. Results of the present study revealed that OTUB2 enhanced the OPG/RANKL ratio both in vivo and in vitro, leading to an increase in bone mass and ossification.

Fracture healing is a complex and long-term process involving callus formation and multiple dynamic stages. The initial stage involves a hematoma, which generates an inflammatory environment. In addition, middle-to-late-stage fracture healing involves endochondral ossification and removal and calcification of the endochondral cartilage. The final stage of bone fracture healing involves chronic remodeling (9). Results of the pathological analysis demonstrated that a few calcified cartilages and minimal levels of uncalcified cartilage existed in the fracture calluses of rats with OTUB2 overexpression at 8 weeks post-fracture. Notably, the cartilage callus had undergone ossification and woven bone had formed. Comparable results were obtained using in vitro experiments. In addition, bone formation depends on the amount and activity of osteoblasts during bone remodeling, which are differentiated from osteoprogenitor cells and BMSCs. The osteogenic differentiation of BMSCs involves pre-osteoblasts, osteoblasts, mature osteoblasts and the deposition and mineralization of the extracellular matrix (44). Several factors have been found to regulate BMSC osteogenic differentiation. Of these, RUNX2 serves an essential role. The onset of osteogenic differentiation is characterized by the increased expression of the RUNX2 protein (45,46). Results of a previous study reveal that RUNX2 induces TNAP activity (45), which promotes bone mineralization (47). Notably, the loss of OTUB2 may inhibit TNAP activity and reduce RUNX2 expression during the osteogenesis of BMSCs (18). These results are comparable with those obtained in the present study. The present study found that OTUB2 may promote the osteogenic differentiation of BMSCs via the facilitation of TNAP activity and upregulation of RUNX2 expression.

Results of the present study revealed that OTUB2 overexpression accelerates fracture healing in vivo and promotes the osteogenic differentiation of BMSCs in vitro. As a downstream protein of OTUB2, it is possible that TRAF3 may exert a similar role in bone repair and formation. As a member of the TRAF family, TRAF3 serves a crucial role in the development of an immune response (48,49). Results of previous studies revealed that TRAF3 may promote bone formation and bone remodeling and reduce bone destruction (41,50). In addition, Yao et al (51) reveal that TRAF3 knockdown in myeloid cells inhibits bone formation in a mouse osteoporosis model. A previous study demonstrates that TRAF3 knockdown in mesenchymal progenitor cells leads to the early onset of osteoporosis in mice, due to decreased bone formation and enhanced bone destruction. Collectively, these results demonstrate that TRAF3 positively regulates the differentiation of mesenchymal progenitor cells into osteoblasts and promotes osteogenesis (49). In addition, TRAF3 overexpression facilitates the osteogenic differentiation and suppresses the adipocytic differentiation of rat BMSCs (50). Results of previous studies also demonstrate that increased TRAF3 expression mediates the inhibition of osteoclastogenesis (5254). Collectively, these results highlight the therapeutic potential of TRAF3 in bone-regulated diseases. Results of the present study revealed that downregulation of TRAF3 repressed the osteogenic differentiation and mineralization of BMSCs, which are the key process in bone healing. Based on results of the OTUB2-induced deubiquitination of TRAF3, it was hypothesized that the protective role of OTUB2 was at least partly mediated by the deubiquitination and accumulation of TRAF3. It is possible that TRAF3 might also act as a part in bone fracture healing. Lack of verifying experiments is a limitation of the present study.

Results of the co-immunoprecipitation analysis demonstrated that OTUB2 interacted with TRAF3. Deubiquitinases possess ubiquitin-binding sites that guide the ubiquitin C terminus and the scissile bond into the active site for hydrolysis (55). Thus, it was hypothesized that the interaction between TRAF3 and OTUB2 may be associated with the deubiquitination of TRAF3. Results of the present study revealed that OTUB2 overexpression reduced the ubiquitination of TRAF3. To further determine whether OTUB2 directly deubiquitinates TRAF3, HEK293 cells expressing inactive enzyme mutant OTUB2 C51S were generated. Results of the co-immunoprecipitation analysis revealed that the OTUB2 mutation reversed the OTUB2 wild-type mediated deubiquitination of TRAF3, indicating that OTUB2 repressed TRAF3 ubiquitination and this was dependent on its deubiquitinase activity. Thus, results of the present study revealed that OTUB2 may induce the deubiquitination of TRAF3, leading to the accumulation of TRAF3 in cells. Notably, these results are comparable with those of a previous study (56). In addition, results of the present study revealed that TRAF3 knockdown inhibited OTUB2-mediated osteogenic differentiation. Thus, OTUB2-mediated TRAF3 deubiquitination may serve a vital role in the process of bone healing.

In conclusion, results of the present study revealed that OTUB2 may promote bone fracture healing through the deubiquitination of TRAF3. Thus, OTUB2 may exhibit potential as a novel therapeutic target in the treatment of fracture and the use of OTUB2 in clinical practice may improve patient outcomes.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Key R&D Program of the China Ministry of Science and Technology (grant no. 2024YFC2510600) and Natural Science Foundation of Hebei Province (grant no. H2022206432).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

LZ, JG, SF, YuZ, HW, HM, WC, YiZ and ZH conceived and designed the research. LZ performed experiments, wrote the manuscript and obtained funding. JG and SF performed experiments and bioinformatics analysis. YuZ and HW performed data acquisition, analysis and interpretation. HM, WC and YiZ conducted statistical analysis and provided substantial intellectual input during the drafting and revision of the manuscript. ZH oversaw the research program, obtained funding and reviewed the manuscript. LZ and ZH confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Animal experiments were approved by Laboratory Animal Ethical and Welfare Committee of Hebei Medical University (approval no. IACUC-Hebmu-2021007) following The Guideline for the Care and Use of Laboratory Animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BMSCs

bone marrow mesenchymal stem cells

OTUB2

otubain 2

OPG

osteoprotegerin

RANKL

receptor activator of nuclear factor-kappa B ligand

RUNX2

runt related transcription factor 2

TNAP

tissue-nonspecific alkaline phosphatase

TRAF3

TNF-receptor associated factor 3

References

1 

Marsell R and Einhorn TA: The biology of fracture healing. Injury. 42:551–555. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Singh V: Medicinal plants and bone healing. Natl J Maxillofac Surg. 8:4–11. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Quirk BJ, Sannagowdara K, Buchmann EV, Jensen ES, Gregg DC and Whelan HT: Effect of near-infrared light on in vitro cellular ATP production of osteoblasts and fibroblasts and on fracture healing with intramedullary fixation. J Clin Orthop Trauma. 7:234–241. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Cheng TL, Schindeler A and Little DG: BMP-2 delivered via sucrose acetate isobutyrate (SAIB) improves bone repair in a rat open fracture model. J Orthop Res. 34:1168–1176. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Praemer A, Furner SE and Rice DP: Musculoskeletal conditions in the United States. Am Acad Orthop Surg. 22:1–199. 1976.

6 

Antapur P, Mahomed N and Gandhi R: Fractures in the elderly: When is hip replacement a necessity? Clin Interv Aging. 6:1–7. 2011.PubMed/NCBI

7 

Giannoudis PV, Einhorn TA and Marsh D: Fracture healing: The diamond concept. Injury. 38 (Suppl 4):S3–S6. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Zhang L, Jin L, Guo J, Bao K, Hu J, Zhang Y, Hou Z and Zhang L: Chronic intermittent hypobaric hypoxia enhances bone fracture healing. Front Endocrinol (Lausanne). 11:5826702021. View Article : Google Scholar : PubMed/NCBI

9 

Saul D and Khosla S: Fracture healing in the setting of endocrine diseases, aging and cellular senescence. Endocr Rev. 43:984–1002. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Park J, Cho J and Song EJ: Ubiquitin-proteasome system (UPS) as a target for anticancer treatment. Arch Pharm Res. 43:1144–1161. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Li M, Chen D, Shiloh A, Luo J, Nikolaev AY, Qin J and Gu W: Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature. 416:648–653. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Ouyang S, Zeng Z, Liu Z, Zhang Z, Sun J, Wang X, Ma M, Ye X, Yu J and Kang W: OTUB2 regulates KRT80 stability via deubiquitination and promotes tumour proliferation in gastric cancer. Cell Death Discov. 8:452022. View Article : Google Scholar : PubMed/NCBI

13 

Zhu Q, Fu Y, Li L, Liu CH and Zhang L: The functions and regulation of Otubains in protein homeostasis and diseases. Ageing Res Rev. 67:1013032021. View Article : Google Scholar : PubMed/NCBI

14 

Stanišić V, Malovannaya A, Qin J, Lonard DM and O'Malley BW: OTU Domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) deubiquitinates estrogen receptor (ER) alpha and affects ERalpha transcriptional activity. J Biol Chem. 284:16135–16145. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D and Manolagas SC: Estrogens and androgens in skeletal physiology and pathophysiology. Physiol Rev. 97:135–187. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Kim JM, Lin C, Stavre Z, Greenblatt MB and Shim JH: Osteoblast-osteoclast communication and bone homeostasis. Cells. 9:20732020. View Article : Google Scholar : PubMed/NCBI

17 

Jähn K, Saito H, Taipaleenmäki H, Gasser A, Hort N, Feyerabend F, Schlüter H, Rueger JM, Lehmann W, Willumeit-Römer R and Hesse E: Intramedullary Mg2Ag nails augment callus formation during fracture healing in mice. Acta Biomater. 36:350–360. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Li XY, Mao XF, Tang XQ, Han QQ, Jiang LX, Qiu YM, Dai J and Wang YX: Regulation of Gli2 stability by deubiquitinase OTUB2. Biochem Biophys Res Commun. 505:113–118. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Jiang Y, Zhang J, Li Z and Jia G: Bone marrow mesenchymal stem cell-derived exosomal miR-25 regulates the ubiquitination and degradation of Runx2 by SMURF1 to promote fracture healing in mice. Front Med (Lausanne). 7:5775782020. View Article : Google Scholar : PubMed/NCBI

20 

Wang F, Guo J, Wang Y, Hu Y, Zhang H, Chen J, Jing Y, Cao L, Chen X and Su J: Loss of Bcl-3 delays bone fracture healing through activating NF-κB signaling in mesenchymal stem cells. J Orthop Translat. 35:72–80. 2022. View Article : Google Scholar : PubMed/NCBI

21 

Arthur A and Gronthos S: Clinical application of bone marrow mesenchymal stem/stromal cells to repair skeletal tissue. Int J Mol Sci. 21:97592020. View Article : Google Scholar : PubMed/NCBI

22 

Wang X, Wang C, Gou W, Xu X, Wang Y, Wang A, Xu W, Guo Q, Liu S, Lu Q, et al: The optimal time to inject bone mesenchymal stem cells for fracture healing in a murine model. Stem Cell Res Ther. 9:2722018. View Article : Google Scholar : PubMed/NCBI

23 

Chen L, Shi K, Ditzel N, Qiu W, Figeac F, Nielsen LHD, Tencerova M, Kowal JM, Ding M, Andreasen CM, et al: KIAA1199 deficiency enhances skeletal stem cell differentiation to osteoblasts and promotes bone regeneration. Nat Commun. 14:20162023. View Article : Google Scholar : PubMed/NCBI

24 

Jing Z, Qiong Z, Yonggang W and Yanping L: Rat bone marrow mesenchymal stem cells improve regeneration of thin endometrium in rat. Fertil Steril. 101:587–594. 2014. View Article : Google Scholar : PubMed/NCBI

25 

National Research Counci: Committee for the Update of the Guide for the Care and Use of Laboratory Animals, . Guide for the Care and Use of Laboratory Animals. 8th. National Academies Press; Washington, DC: 2011

26 

Song JL, Zheng W, Chen W, Qian Y, Ouyang YM and Fan CY: Lentivirus-mediated microRNA-124 gene-modified bone marrow mesenchymal stem cell transplantation promotes the repair of spinal cord injury in rats. Exp Mol Med. 49:e3322017. View Article : Google Scholar : PubMed/NCBI

27 

Chrastil J, Sampson C, Jones KB and Higgins TF: Postoperative opioid administration inhibits bone healing in an animal model. Clin Orthop Relat Res. 471:4076–4081. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Kelly LS, Munley JA, Pons EE, Kannan KB, Whitley EM, Bible LE, Efron PA and Mohr AM: A rat model of multicompartmental traumatic injury and hemorrhagic shock induces bone marrow dysfunction and profound anemia. Animal Model Exp Med. 7:367–376. 2024. View Article : Google Scholar : PubMed/NCBI

29 

Munley JA, Kelly LS, Park G, Gillies GS, Pons EE, Kannan KB, Whitley EM, Bible LE, Efron PA, Nagpal R and Mohr AM: Multicompartmental traumatic injury induces sex-specific alterations in the gut microbiome. J Trauma Acute Care Surg. 95:30–38. 2023. View Article : Google Scholar : PubMed/NCBI

30 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Liang W, Ding P, Li G, Lu E and Zhao Z: Hydroxyapatite nanoparticles facilitate osteoblast differentiation and bone formation within sagittal suture during expansion in rats. Drug Des Devel Ther. 15:905–917. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Tseng JC, Meganck J, Peterson JD and Hopkinton M: Quantum GX microCT Imaging System: Features and Performance. PerkinElmer; Hopkington, MA: 2015

33 

Ishikawa M, Ito H, Kitaori T, Murata K, Shibuya H, Furu M, Yoshitomi H, Fujii T, Yamamoto K and Matsuda S: MCP/CCR2 signaling is essential for recruitment of mesenchymal progenitor cells during the early phase of fracture healing. PLoS One. 9:e1049542014. View Article : Google Scholar : PubMed/NCBI

34 

Ferrin I, Beloqui I, Zabaleta L, Salcedo JM, Trigueros C and Martin AG: Isolation, culture, and expansion of mesenchymal stem cells. Methods Mol Biol. 1590:177–190. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Sun Y, Xu L, Huang S, Hou Y, Liu Y, Chan KM, Pan XH and Li G: mir-21 overexpressing mesenchymal stem cells accelerate fracture healing in a rat closed femur fracture model. Biomed Res Int. 2015:4123272015. View Article : Google Scholar : PubMed/NCBI

36 

Sun Y, Xu J, Xu L, Zhang J, Chan K, Pan X and Li G: MiR-503 promotes bone formation in distraction osteogenesis through suppressing Smurf1 expression. Sci Rep. 7:4092017. View Article : Google Scholar : PubMed/NCBI

37 

Samuel S, Ahmad RE, Ramasamy TS, Karunanithi P, Naveen SV, Murali MR, Abbas AA and Kamarul T: Platelet-rich concentrate in serum free medium enhances osteogenic differentiation of bone marrow-derived human mesenchymal stromal cells. PeerJ. 4:e23472016. View Article : Google Scholar : PubMed/NCBI

38 

Faul F, Erdfelder E, Lang AG and Buchner A: G*Power 3: A flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav Res Methods. 39:175–191. 2007. View Article : Google Scholar : PubMed/NCBI

39 

De Winter JCF: Using the Student's t-test with extremely small sample sizes. Pract Assess Res Eval. 18:102013.

40 

Sullivan GM and Feinn R: Using effect size-or why the P-value is not enough. J Grad Med Educ. 4:279–282. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Ayoub A, Xiu Y, Yin X, Sanders JO, Mesfin A, Xing L, Yao Z and Boyce BF: TGFβ-induced degradation of TRAF3 in mesenchymal progenitor cells causes age-related osteoporosis. Nat Commun. 10:27952019. View Article : Google Scholar : PubMed/NCBI

42 

Hashimoto K, Shinyashiki Y, Ohtani K, Kakinoki R and Akagi M: How proximal femur fracture patients aged 65 and older fare in survival and cause of death 5+ years after surgery: A long-term follow-up. Medicine (Baltimore). 102:e338632023. View Article : Google Scholar : PubMed/NCBI

43 

Boyce BF and Xing L: The RANKL/RANK/OPG pathway. Curr Osteoporos Rep. 5:98–104. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Ma C, Gao J, Liang J, Dai W, Wang Z, Xia M, Chen T, Huang S, Na J, Xu L, et al: HDAC6 inactivates Runx2 promoter to block osteogenesis of bone marrow stromal cells in age-related bone loss of mice. Stem Cell Res Ther. 12:4842021. View Article : Google Scholar : PubMed/NCBI

45 

Fujita T, Azuma Y, Fukuyama R, Hattori Y, Yoshida C, Koida M, Ogita K and Komori T: Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J Cell Biol. 166:85–95. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Ducy P, Zhang R, Geoffroy V, Ridall AL and Karsenty G: Osf2/Cbfa1: A transcriptional activator of osteoblast differentiation. Cell. 89:747–754. 1997. View Article : Google Scholar : PubMed/NCBI

47 

Haarhaus M, Cianciolo G, Barbuto S, La Manna G, Gasperoni L, Tripepi G, Plebani M, Fusaro M and Magnusson P: Alkaline phosphatase: An old friend as treatment target for cardiovascular and mineral bone disorders in chronic kidney disease. Nutrients. 14:21242022. View Article : Google Scholar : PubMed/NCBI

48 

Chang JH, Hu H, Jin J, Puebla-Osorio N, Xiao Y, Gilbert BE, Brink R, Ullrich SE and Sun SC: TRAF3 regulates the effector function of regulatory T cells and humoral immune responses. J Exp Med. 211:137–151. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Yi Z, Lin WW, Stunz LL and Bishop GA: Roles for TNF-receptor associated factor 3 (TRAF3) in lymphocyte functions. Cytokine Growth Factor Rev. 25:147–156. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Wang D, Cai G, Wang H and He J: TRAF3, a target of MicroRNA-363-3p, suppresses senescence and regulates the balance between osteoblastic and adipocytic differentiation of rat bone marrow-derived mesenchymal stem cells. Stem Cells Dev. 29:737–745. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Yao Z, Ayoub A, Srinivasan V, Wu J, Tang C, Duan R, Milosavljevic A, Xing L, Ebetino FH, Frontier AJ and Boyce BF: Hydroxychloroquine and a low antiresorptive activity bisphosphonate conjugate prevent and reverse ovariectomy-induced bone loss in mice through dual antiresorptive and anabolic effects. Bone Res. 12:522024. View Article : Google Scholar : PubMed/NCBI

52 

Yao Z, Lei W, Duan R, Li Y, Luo L and Boyce BF: RANKL cytokine enhances TNF-induced osteoclastogenesis independently of TNF receptor associated factor (TRAF) 6 by degrading TRAF3 in osteoclast precursors. J Biol Chem. 292:10169–10179. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Boyce BF, Li J, Xing L and Yao Z: Bone remodeling and the role of TRAF3 in osteoclastic bone resorption. Front Immunol. 9:22632018. View Article : Google Scholar : PubMed/NCBI

54 

Xiu Y, Xu H, Zhao C, Li J, Morita Y, Yao Z, Xing L and Boyce BF: Chloroquine reduces osteoclastogenesis in murine osteoporosis by preventing TRAF3 degradation. J Clin Invest. 124:297–310. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Mevissen TET and Komander D: Mechanisms of deubiquitinase specificity and regulation. Annu Rev Biochem. 86:159–192. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Li S, Zheng H, Mao AP, Zhong B, Li Y, Liu Y, Gao Y, Ran Y, Tien P and Shu HB: Regulation of virus-triggered signaling by OTUB1- and OTUB2-mediated deubiquitination of TRAF3 and TRAF6. J Biol Chem. 285:4291–4297. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 32 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Guo J, Feng S, Zheng Y, Wang H, Ma H, Chen W, Zhang Y and Hou Z: Otubain 2 stabilizes TNF‑receptor associated factor 3 to accelerate bone fracture healing. Mol Med Rep 32: 223, 2025.
APA
Zhang, L., Guo, J., Feng, S., Zheng, Y., Wang, H., Ma, H. ... Hou, Z. (2025). Otubain 2 stabilizes TNF‑receptor associated factor 3 to accelerate bone fracture healing. Molecular Medicine Reports, 32, 223. https://doi.org/10.3892/mmr.2025.13588
MLA
Zhang, L., Guo, J., Feng, S., Zheng, Y., Wang, H., Ma, H., Chen, W., Zhang, Y., Hou, Z."Otubain 2 stabilizes TNF‑receptor associated factor 3 to accelerate bone fracture healing". Molecular Medicine Reports 32.2 (2025): 223.
Chicago
Zhang, L., Guo, J., Feng, S., Zheng, Y., Wang, H., Ma, H., Chen, W., Zhang, Y., Hou, Z."Otubain 2 stabilizes TNF‑receptor associated factor 3 to accelerate bone fracture healing". Molecular Medicine Reports 32, no. 2 (2025): 223. https://doi.org/10.3892/mmr.2025.13588