1
|
Zhang Z, Cao J, Xing H, Liu J, Li L, Zhang Y. USP2-induced upregulation of LEF1 through deubiquitination relieves osteoporosis development by promoting the osteogenic differentiation of bone marrow mesenchymal stem cells. J Orthop Surg Res 2025; 20:430. [PMID: 40301985 PMCID: PMC12039128 DOI: 10.1186/s13018-025-05834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) exhibit therapeutic potential for osteoporosis through their differentiation into osteoblasts. Here, we investigated the role and mechanism of lymphoid enhancer-binding factor 1 (LEF1) in regulating osteogenic differentiation of human BMSCs (hBMSCs). METHODS hBMSCs were exposed to the specific medium to induce their osteogenic differentiation. The ovariectomy (OVX)-induced osteoporotic mouse model was constructed. LEF1 and USP2 mRNA expression was analyzed by quantitative PCR, and protein levels were detected by immunohistochemistry and immunoblotting. Cell proliferation was assessed by CCK-8 assay. Alkaline phosphatase (ALP) expression and activity assay and Alizarin Red staining were used to evaluate osteogenic differentiation. LEF1 protein stability analysis and co-immunoprecipitation (Co-IP) assay were performed to test the USP2/LEF1 interaction. RESULTS During hBMSC osteogenic differentiation, LEF1 and USP2 levels were increased in hBMSCs. Inhibiting LEF1 or USP2 diminished the proliferation and osteogenic differentiation of hBMSCs in vitro. Mechanistically, USP2 stabilized LEF1 protein by mediating LEF1 deubiquitination. Increased expression of LEF1 reversed USP2 knockdown-imposed suppression on proliferation and osteogenic differentiation of hBMSCs. Moreover, increased USP2 expression reduced bone loss and enhanced osteogenic differentiation in OVX mice. Additionally, LEF1 and USP2 were downregulated in the bone marrow of patients with osteoporosis. CONCLUSION Our findings provide the first demonstration of the USP2/LEF1 cascade that enhances the osteogenic differentiation of hBMSCs, broadening the field for the development of BMSCs as effective agents in osteoporosis therapy.
Collapse
Affiliation(s)
- Zhihong Zhang
- Department of Orthopedics Ward 2, Kailuan General Hospital, No. 57, Xinhua East Road, Tangshan City, Hebei Province, 063000, China
| | - Jie Cao
- Department of Orthopedics Ward 2, Kailuan General Hospital, No. 57, Xinhua East Road, Tangshan City, Hebei Province, 063000, China
| | - Hanwen Xing
- Department of Hematology, Kailuan General Hospital, Tangshan City, Hebei, 063000, China
| | - Jing Liu
- Department of Operating Room, Tangshan people's hospital Guye Institute District, Tangshan City, Hebei, 063000, China
| | - Linshuo Li
- Department of Orthopedics Ward 2, Kailuan General Hospital, No. 57, Xinhua East Road, Tangshan City, Hebei Province, 063000, China
| | - Yue Zhang
- Department of Orthopedics Ward 2, Kailuan General Hospital, No. 57, Xinhua East Road, Tangshan City, Hebei Province, 063000, China.
| |
Collapse
|
2
|
Dvir-Ginzberg M, Maatuf YH, Mobasheri A. Do we understand sex-related differences governing dimorphic disease mechanisms in preclinical animal models of osteoarthritis? Osteoarthritis Cartilage 2024; 32:1054-1057. [PMID: 38531464 DOI: 10.1016/j.joca.2024.03.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
Research conducted using murine preclinical models of osteoarthritis (OA) over the last three decades has brought forth many exciting developments showcasing mechanisms and pathways that drive disease pathogenesis. These models have identified therapeutic targets that can be modulated via innovative biologicals and pharmaceuticals. However, many of these approaches have failed to translate to humans and reach the clinic. This commentary aims to highlight some of the key hurdles in the translation of novel findings using preclinical OA models with a focus on sex-related differences and variations in chondrosenescence in these animal models. Notably, besides chondrosenescence, other signaling mechanisms have been shown to be affected by sexual dimorphism (i.e. TGFβ signaling, EGFR/integrin α1β1 and Trpv4). Preclinical models of OA mainly utilize male mice due to their capacity to manifest fast progressing OA structural phenotype compared to female mice. This experimental trend has overlooked and ignored the sex-related effects of numerous mechanisms affecting joint structure, that influence OA structural progression. Future work should focus on analyzing both sexes and understanding sex-related differences, which will enable us to gain a better understanding of the progression of OA based on sex-related mechanistic discrepancies, and potentially improve translatability.
Collapse
Affiliation(s)
- Mona Dvir-Ginzberg
- Laboratory of Cartilage Biology, Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Yonathan H Maatuf
- Laboratory of Cartilage Biology, Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
3
|
Maatuf YH, Marco M, Unger-Gelman S, Farhat E, Zobrab A, Roy A, Kumar A, Carmon I, Reich E, Dvir-Ginzberg M. Diverse Response to Local Pharmacological Blockade of Sirt1 Cleavage in Age-Induced versus Trauma-Induced Osteoarthritis Female Mice. Biomolecules 2024; 14:81. [PMID: 38254681 PMCID: PMC10813022 DOI: 10.3390/biom14010081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Objective: Previous studies have shown that the cleavage of Sirt1 contributes to the development of osteoarthritis (OA). In fact, OA was effectively abrogated by the intra-articular (IA) administration of two compounds, one blocking Sirt1 cleavage (CA074me) and the other activating Sirt1 (SRT1720), using a post-traumatically induced model (PTOA) in young female mice. In this study, we attempted to understand if this local treatment is effective in preventing age-associated OA (AOA) progression and symptoms. Design: A group of 17-month-old female C57BL/6J mice were IA administered with CA074me and/or SRT1720 or their combination. Joint histopathological analysis and bone histomorphometry were carried out, with an assessment of knee mechanical hyperalgesia. A serum analysis for NT/CT Sirt1 was carried out along with immunohistochemistry for articular cartilage to detect p16INK4A or γH2A.X. Similarly, meniscal cartilage was monitored for Lef1 and Col1a1 deposition. The data were compared for young female mice subjected to post-traumatic OA (PTOA). Results: Similar to PTOA, combination-treated AOA exhibited improved knee hyperalgesia, yet structural improvements were undetected, corresponding to unchanged NT/CT Sirt1 serum levels. Both AOA and PTOA exhibited unchanged staining for nuclear p16INK4A or γH2A.X and lacked a correlation with OA severity. Contrarily to PTOA, the combination treatment with AOA did not exhibit a local reduction in the Lef1 and Col1 targets. Conclusions: When targeting Sirt1 cleavage, the PTOA and AOA models exhibited a similar pain response to the combination treatment; however, they displayed diverse structural outcomes for joint-related damage, related to Lef1-dependent signaling. Interestingly, nuclear p16INK4A was unaffected in both models, regardless of the treatment's effectiveness. Finally, these findings highlight the variations in the responses between two highly researched OA preclinical models, reflecting OA pathophysiology heterogeneity and variations in gender-related drug-response mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mona Dvir-Ginzberg
- Institute of Biomedical and Oral research, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 9112102, Israel; (Y.H.M.); (M.M.); (S.U.-G.); (E.F.); (A.Z.); (A.R.); (A.K.); (I.C.); (E.R.)
| |
Collapse
|
4
|
Ackert-Bicknell C, Karasik D. Proceedings of the Post-Genome Analysis for Musculoskeletal Biology Workshop. Curr Osteoporos Rep 2023; 21:184-192. [PMID: 36869984 DOI: 10.1007/s11914-023-00781-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/05/2023]
Abstract
PURPOSE OF THE REVIEW Herein, we report on the proceedings of the workshop entitled "Post-Genome analysis for musculoskeletal biology" that was held in July of 2022 in Safed, Galilee, Israel. Supported by the Israel Science Foundation, the goal of this workshop was to bring together established investigators and their trainees who were interested in understanding the etiology of musculoskeletal disease, from Israel and from around the world. RECENT FINDINGS Presentations at this workshop spanned the spectrum from basic science to clinical studies. A major emphasis of the discussion centered on genetic studies in humans, and the limitations and advantages of such studies. The power of coupling studies using human data with functional follow-up studies in pre-clinical models such as mice, rats, and zebrafish was discussed in depth. The advantages and limitations of mice and zebrafish for faithfully modelling aspects of human disease were debated, specifically in the context of age-related diseases such as osteoporosis, osteoarthritis, adult-onset auto-immune disease, and osteosarcopenia. There remain significant gaps in our understanding of the nature and etiology of human musculoskeletal disease. While therapies and medications exist, much work is still needed to find safe and effective interventions for all patients suffering from diseases associated with age-related deterioration of musculoskeletal tissues. The potential of forward and reverse genetic studies has not been exhausted for diseases of muscles, joints, and bones.
Collapse
Affiliation(s)
- Cheryl Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado, Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO, 80045, USA.
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
5
|
Yu Z, Zhang Z, Zhang X, Bao J, Li H, Yu J, Shi N, Nan F, Cao L, Li C, Wang W. 4-Octyl itaconate treatment inhibits mitochondrial dysfunction and ER stress via stimulating SIRT1 expression in vitro and attenuates osteoarthritis process in murine DMM model in vivo. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
|
6
|
Carmon I, Kalmus S, Zobrab A, Alterman M, Emram R, Gussarsky M, Kandel L, Reich E, Casap N, Dvir-Ginzberg M. Repairing a critical cranial defect using WISP1-pretreated chondrocyte scaffolds. J Tissue Eng 2023; 14:20417314231159740. [PMID: 36949842 PMCID: PMC10026108 DOI: 10.1177/20417314231159740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/09/2023] [Indexed: 03/19/2023] Open
Abstract
In cranial flat bone fractures, spontaneous bone repair will occur only when the fracture ends are in close contact. However, in cases wherein bone discontinuity is extensive, surgical interventions are often required. To this end, autologous bone is harvested and surgically integrated into the site of fracture. Here we propose to use cartilage, as an alternative autologous source, to promote cranial fracture repair. The advantage of this approach is the potential reduction in donor site morbidity, likely due to the avascular and aneural nature of cartilage. As a first step we attempted to induce cartilage mineralization in vitro, using micromass primary chondrocyte cultures, incubated with BMP2 and/or WISP1, which were examined histologically following a 3-week culture period. Next, chondrocyte seeded collagen scaffolds were evaluated in vitro for expression profiles and ALP activity. Finally, chondrocyte-seeded collagen scaffolds were implanted in a Lewis rats 8 mm critical calvaria defect model, which was imaged via live CT for 12 weeks until sacrifice. End points were analyzed for microCT, histology, and serum levels of bone related markers. Micromass cultures exhibited an osseous inducing trend following WISP1 administration, which was maintained in chondrocyte seeded scaffolds. Accordingly, in vivo analysis was carried out to assess the impact of WISP1-pretreated chondrocytes (WCS) versus untreated chondrocytes (UCS) in calvaria defect model and compared to untreated control comprised of a defect-associated blood clot (BC) or empty collagen scaffold (CS) implant. Live CT and microCT exhibited higher mineralization volumes in critical defect implanted with UCS, with some structural improvements in WCS. Histological analysis exhibited higher anabolic bone formation in WCS and trabecular bone was detected in WCS and UCS groups. Chondrocytes implanted into critical cranial defect expedite the formation of native-like osseous tissue, especially after WISP1 priming in culture. Ultimately, these data support the use of autologous chondrocytes to repair critical maxillofacial defects.
Collapse
Affiliation(s)
- Idan Carmon
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Shira Kalmus
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Anna Zobrab
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Michael Alterman
- Deptatement. of Maxillofacial Surgery,
Faculty of Dental Medicine, Hadassah-Hebrew University, Jerusalem, Israel
| | - Raphaelle Emram
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - May Gussarsky
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Leonid Kandel
- Orthopedic Research Unit,
Hadassah-Hebrew University, Jerusalem, Israel
| | - Eli Reich
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Nardi Casap
- Deptatement. of Maxillofacial Surgery,
Faculty of Dental Medicine, Hadassah-Hebrew University, Jerusalem, Israel
| | - Mona Dvir-Ginzberg
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
- Mona Dvir-Ginzberg, Laboratory of Cartilage
Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine,
Hebrew University of Jerusalem, P. O. Box 12272, Jerusalem 9112102, Israel.
| |
Collapse
|
7
|
Carmon I, Zecharyahu L, Elayyan J, Meka SRK, Reich E, Kandel L, Bilkei-Gorzo A, Zimmer A, Mechoulam R, Kravchenko-Balasha N, Dvir-Ginzberg M. HU308 Mitigates Osteoarthritis by Stimulating Sox9-Related Networks of Carbohydrate Metabolism. J Bone Miner Res 2023; 38:154-170. [PMID: 36350089 PMCID: PMC10098743 DOI: 10.1002/jbmr.4741] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Osteoarthritis (OA) is characterized by progressive, irreversible erosion of articular cartilage accompanied by severe pain and immobility. This study aimed to assess the effect and mechanism of action of HU308, a selective cannabinoid receptor type 2 (CB2) agonist, in preventing OA-related joint damage. To test the assumption that HU308 could prevent OA-related joint damage, Cnr2 null mice and wild type (WT) mice were aged to reach 20 months and analyzed for joint structural features. OA was induced in WT mice via a post-traumatic procedure or aging, followed by HU308 local (intra-articular) or systemic (intraperitoneal) administration, respectively. Additional analyses of time and dose courses for HU308 were carried out in human primary chondrocytes, analyzed by RNA sequencing, RT-PCR, chromatin immunoprecipitation, and immunoblotting. Our results showed that Cnr2 null mice exhibited enhanced age-related OA severity and synovitis compared to age-matched WT mice. Systemic administration of HU308 to 16-month-old mice improved pain sensitivity and maintained joint integrity, which was consistent with the intra-articular administration of HU308 in post-traumatic OA mice. When assessing human chondrocytes treated with HU308, we uncovered a dose- and time-related increase in ACAN and COL2A1 expression, which was preceded by increased SOX9 expression due to pCREB transcriptional activity. Finally, transcriptomic analysis of patient-derived human chondrocytes identified patient subpopulations exhibiting HU308-responsive trends as judged by enhanced SOX9 expression, accompanied by enriched gene networks related to carbohydrate metabolism. Collectively, the results showed that HU308 reduced trauma and age-induced OA via CB2-pCREB dependent activation of SOX9, contributing to augmented gene networks related to carbohydrate metabolism. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Idan Carmon
- Multidisciplinary Center for Cannabinoid Research, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lital Zecharyahu
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jinan Elayyan
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sai R K Meka
- Multidisciplinary Center for Cannabinoid Research, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eli Reich
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leonid Kandel
- Orthopedic Complex. Hebrew University- Hadassah Medical Center, Jerusalem, Israel
| | | | - Andreas Zimmer
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Raphael Mechoulam
- Multidisciplinary Center for Cannabinoid Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nataly Kravchenko-Balasha
- Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mona Dvir-Ginzberg
- Multidisciplinary Center for Cannabinoid Research, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of BioMedical and Oral Research, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
8
|
Li Q, Xu P, Zhang C, Gao Y. MiR-362-5p inhibits cartilage repair in osteoarthritis via targeting plexin B1. J Orthop Surg (Hong Kong) 2022; 30:10225536221139887. [PMID: 36523183 DOI: 10.1177/10225536221139887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Chondrogenesis of bone marrow mesenchymal stem cells (BMSCs) exerts great function during the pathogenesis of osteoarthritis (OA). Studies have reported the association of plexin B1 (PLXNB1) with OA pathogenesis. In this study, the upstream mechanism and function of PLXNB1 in this disease were explored. METHODS Flow cytometry was applied to test BMSC characterization. Chondrogenic differentiation of BMSCs was evaluated by Alcian blue staining. The expression of PLXNB1, miR-362-5p, miR-501-5p, miR-1827, miR-500-5p was measured using RT-qPCR analysis. The protein levels of PLXNB1, Aggrecan, and Silent information regulator factor 2-related enzyme 1 (SIRT1) were determined by western blotting. Binding relationship between miR-362-5p and PLXNB1 was confirmed using bioinformatics analysis and luciferase reporter assay. The in vivo model of OA was established in Sprague-Dawley rats which received medial meniscus instability surgery. For histopathological examination, cartilage tissues in the knee joint of rats were stained with hematoxylin and eosin. Micro-CT analysis was employed to observe the changes of morphometric indices including average trabecular separation, average trabecular thickness, and bone volume fraction. RESULTS BMSCs were identified to possess the characteristics of mesenchymal stem cells. PLXNB1 was observed to be highly expressed during chondrogenic differentiation of BMSCs and PLXNB1 overexpression promoted BMSC chondrogenic differentiation. Mechanically, PLXNB1 was targeted by miR-362-5p. In rescue assays, miR-362-5p reversed the effects of PLXNB1 on chondrogenic differentiation of BMSCs. In the in vivo experiments, upregulated PLXNB1 expression alleviated joint injury of OA rats. Additionally, overexpressed miR-362-5p and downregulated PLXNB1 expression levels were detected in OA rats. CONCLUSION MiR-362-5p promotes OA progression by suppressing PLXNB1.
Collapse
Affiliation(s)
- Qian Li
- Department of Massage, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Ping Xu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Chi Zhang
- Department of Massage, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Yang Gao
- Department of Massage, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| |
Collapse
|