1
|
Huang D, Li Z, Li G, Zhou F, Wang G, Ren X, Su J. Biomimetic structural design in 3D-printed scaffolds for bone tissue engineering. Mater Today Bio 2025; 32:101664. [PMID: 40206144 PMCID: PMC11979411 DOI: 10.1016/j.mtbio.2025.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
The rising prevalence of bone diseases in an aging population underscores the urgent need for innovative and clinically translatable solutions in bone tissue engineering. While significant progress has been made in refining the chemical properties of biomaterials, the structural design of scaffolds-a critical determinant of repair success-remains comparatively underexplored. Structural parameters such as porosity, pore size, and interconnectivity are not only essential for achieving mechanical stability but also pivotal in regulating biological processes, including vascularization, osteogenesis, and immune modulation. This review systematically categorizes scaffold architectures documented in the literature and highlights how these design parameters can be optimized to enhance bone regeneration. Advanced fabrication technologies, particularly 3D printing, are emphasized for their transformative potential in creating precise, biomimetic scaffolds that align with the complex functional demands of native bone. Furthermore, this work synthesizes diverse findings to provide a comprehensive framework for designing next-generation scaffolds. By bridging the gap between structural innovation and clinical application, this review delivers actionable strategies and a strategic roadmap for advancing the field toward improved clinical outcomes and transformative breakthroughs in regenerative medicine.
Collapse
Affiliation(s)
- Dan Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zuhao Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
2
|
Wang Y, Jan H, Zhong Z, Zhou L, Teng K, Chen Y, Xu J, Xie D, Chen D, Xu J, Qin L, Tuan RS, Li ZA. Multiscale metal-based nanocomposites for bone and joint disease therapies. Mater Today Bio 2025; 32:101773. [PMID: 40290898 PMCID: PMC12033929 DOI: 10.1016/j.mtbio.2025.101773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Bone and joint diseases are debilitating conditions that can result in significant functional impairment or even permanent disability. Multiscale metal-based nanocomposites, which integrate hierarchical structures ranging from the nanoscale to the macroscale, have emerged as a promising solution to this challenge. These materials combine the unique properties of metal-based nanoparticles (MNPs), such as enzyme-like activities, stimuli responsiveness, and photothermal conversion, with advanced manufacturing techniques, such as 3D printing and biohybrid systems. The integration of MNPs within polymer or ceramic matrices offers a degree of control over the mechanical strength, antimicrobial efficacy, and the manner of drug delivery, whilst concomitantly promoting the processes of osteogenesis and chondrogenesis. This review highlights breakthroughs in stimulus-responsive MNPs (e.g., photo-, magnetically-, or pH-activated systems) for on-demand therapy and their integration with biocomposite hybrids containing cells or extracellular vesicles to mimic the native tissue microenvironment. The applications of these composites are extensive, ranging from bone defects, infections, tumors, to degenerative joint diseases. The review emphasizes the enhanced load-bearing capacity, bioactivity, and tissue integration that can be achieved through hierarchical designs. Notwithstanding the potential of these applications, significant barriers to progress persist, including challenges related to long-term biocompatibility, regulatory hurdles, and scalable manufacturing. Finally, we propose future directions, including machine learning-guided design and patient-specific biomanufacturing to accelerate clinical translation. Multiscale metal-based nanocomposites, which bridge nanoscale innovations with macroscale functionality, are a revolutionary force in the field of biomedical engineering, providing personalized regenerative solutions for bone and joint diseases.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Hasnain Jan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Zheng Zhong
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Kexin Teng
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Ye Chen
- Department of Chemistry, Faculty of Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Dexin Chen
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou, 510632, China
| | - Jiake Xu
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, NT, Hong Kong Special Administrative Region of China
| |
Collapse
|
3
|
Bumedien-Abdelgani H, Manzano-Moreno FJ, González-Acedo A, García-Recio E, Ruiz C, de Luna-Bertos E. Effect of clindamycin on human osteoblasts treated with zoledronate: An in vitro study. Arch Oral Biol 2025; 174:106247. [PMID: 40203484 DOI: 10.1016/j.archoralbio.2025.106247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVE The objective of this study was to determine the effects of combined treatment with clindamycin and zoledronate on the growth and differentiation of cultured human osteoblasts. DESIGN Human osteoblasts, obtained by primary culture from mandibular bone fragments, were cultured in the presence of 50 μM zoledronate, 150 μg/mL clindamycin, or the combination of both (zoledronate + clindamycin). The effect on cell proliferation was evaluated at 24 h by the MTT colorimetric method, using a spectrophotometer at 570 nm. The effect on differentiation was examined by measuring alkaline phosphatase (ALP) activity, and mineralization by the osteoblast was studied by staining with alizarin red. Real-time polymerase chain reaction (RT-PCR) was performed for gene expression analysis. Data were expressed as means±standard deviation, and analysis of variance was performed, applying Bonferroni correction when interactions were significant. RESULTS Treatment of osteoblasts with 50 μM zoledronate significantly reduced cell proliferation and differentiation and the gene expression of certain markers versus controls (p < 0.001). However, treatment with 150 μg/mL clindamycin significantly increased cell proliferation and differentiation and the gene expression of certain markers (p < 0.05). The combination of 150 μg/mL clindamycin and 50 μM zoledronate partially counteracted the loss of osteoblast proliferative and differentiation capacity caused by zoledronate. CONCLUSION Treatment with low-dose clindamycin can reverse the negative impact of zoledronate on osteoblast proliferation and differentiation. Follow-up animal studies and clinical trials are needed before topical clindamycin can be considered as a possible therapeutic resource for BP-treated patients who require a GBR procedure.
Collapse
Affiliation(s)
| | - Francisco Javier Manzano-Moreno
- Department of Stomatology, School of Dentistry, University of Granada, Spain; Biomedical Group (BIO277), University of Granada, Spain; Instituto Investigación Biosanitaria, ibs.Granada Spain.
| | - Anabel González-Acedo
- Biomedical Group (BIO277), University of Granada, Spain; Instituto Investigación Biosanitaria, ibs.Granada Spain; Department of Nursing, Faculty of Health Science, University of Granada, Melilla, Spain
| | - Enrique García-Recio
- Biomedical Group (BIO277), University of Granada, Spain; Instituto Investigación Biosanitaria, ibs.Granada Spain; Department of Nursing, Faculty of Health Science, University of Granada, Melilla, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), University of Granada, Spain; Instituto Investigación Biosanitaria, ibs.Granada Spain; Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Elvira de Luna-Bertos
- Biomedical Group (BIO277), University of Granada, Spain; Instituto Investigación Biosanitaria, ibs.Granada Spain; Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| |
Collapse
|
4
|
Jiang J, Wang J, Fan P, Zhao Z, Deng H, Li J, Wang Y, Wang Y. Biomaterial-based strategies for bone cement: modulating the bone microenvironment and promoting regeneration. J Nanobiotechnology 2025; 23:343. [PMID: 40361125 PMCID: PMC12070552 DOI: 10.1186/s12951-025-03363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoporotic bone defect and fracture healing remain significant challenges in clinical practice. While traditional therapeutic approaches provide some regulation of bone homeostasis, they often present limitations and adverse effects. In orthopedic procedures, bone cement serves as a crucial material for stabilizing osteoporotic bone and securing implants. However, with the exception of magnesium phosphate cement, most cement variants lack substantial bone regenerative properties. Recent developments in biomaterial science have opened new avenues for enhancing bone cement functionality through innovative modifications. These advanced materials demonstrate promising capabilities in modulating the bone microenvironment through their distinct physicochemical properties. This review provides a systematic analysis of contemporary biomaterial-based modifications of bone cement, focusing on their influence on the bone healing microenvironment. The discussion begins with an examination of bone microenvironment pathology, followed by an evaluation of various biomaterial modifications and their effects on cement properties. The review then explores regulatory strategies targeting specific microenvironmental elements, including inflammatory response, oxidative stress, osteoblast-osteoclast homeostasis, vascular network formation, and osteocyte-mediated processes. The concluding section addresses current technical challenges and emerging research directions, providing insights for the development of next-generation biomaterials with enhanced functionality and therapeutic potential.
Collapse
Affiliation(s)
- Jiawei Jiang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Juan Wang
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, Jiangsu, China
| | - Pan Fan
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhe Zhao
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China
| | - Hongjian Deng
- Department of Orthopaedics, The Affiliated 2 Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian Li
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| | - Yi Wang
- Department of Orthopaedics, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| | - Yuntao Wang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| |
Collapse
|
5
|
G P, Verma RK, Shukla R. Leveraging Nanoscience and Strategic Delivery for the Expedition of Osteoporosis. AAPS PharmSciTech 2025; 26:129. [PMID: 40341672 DOI: 10.1208/s12249-025-03120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/17/2025] [Indexed: 05/10/2025] Open
Abstract
Osteoporosis is a globally affecting bone disease characterized by reduced bone mineral density, in which women are more insidious to the disease. It accounts for 8.9 million fractures annually, and about 50% of repeated hip fractures cause permanent disabilities. With the knowledge of determinants and pathology, various FDA-approved drugs and therapies are available for the management of the disease, but the challenges associated with those therapies lead to the adoption of nanotechnology in osteoporosis management. The nanosystems developed for the management of osteoporosis are nanogenerators, nanobubbles, microneedles, nanogels, implantable delivery systems, nanoparticles, nanofibrous scaffolds, and nanocements that probably address the current challenges related to the diagnosis and cure. In view of targeted accumulation of the cargo, various moieties assisted the nanocarrier system for selective distribution to bone, and the development of different types of nanotechnology-based delivery systems has been briefed in this review.
Collapse
Affiliation(s)
- Pramoda G
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Rahul K Verma
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab, 160062, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
6
|
Lee SH, Kim M, Jung KI, Lee SJ, Park MH. Ishophloroglucin A Isolated From Ishige okamurae Stimulates Osteoblast Differentiation Through Activation of the Bone Morphogenetic Protein and Wnt/β-Catenin Signaling Pathways in MC3T3-E1 Cells. Cell Biol Int 2025. [PMID: 40343669 DOI: 10.1002/cbin.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/31/2025] [Accepted: 04/26/2025] [Indexed: 05/11/2025]
Abstract
Current osteoporosis treatments are insufficient as they cause a relatively small increase in bone mass and are unable to recover lost bone structures, in addition to having severe side effects. The bone morphogenetic protein (BMP) and Wnt/β-catenin signaling pathways cooperatively modulate bone formation and osteoblast differentiation and therefore may play a role in treating osteoporosis. This study aimed to investigate the effects of Ishophloroglucin A (IPA), a novel phenolic compound isolated from Ishige okamurae, on osteoblast differentiation by activating the BMP and Wnt/β-catenin signaling pathways. According to our findings, IPA significantly promoted the osteogenic proliferation of MC3T3-E1 osteoblastic cells and increased alkaline phosphatase (ALP) activity and calcium nodule formation in MC3T3-E1 cells compared to the untreated control. IPA also upregulated osteogenesis markers such as type 1 collagen, ALP, p-Smad1/5/8, osterix, osteopontin, runt-related transcription factors (Runx2), and BMP2 in MC3T3-E1 cells in a dose-dependent manner. Moreover, IPA activated Wnt3a, LRP5, DVL2, and β-catenin in MC3T3-E1 cells. Overall, our results demonstrate that IPA promotes the differentiation of MC3T3-E1 osteoblastic cells by activating the BMP and Wnt/β-catenin signaling pathways, suggesting that it may be a potential candidate target for treating or preventing osteoporosis.
Collapse
Affiliation(s)
- Seung-Hong Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Mihyang Kim
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan, Republic of Korea
| | - Kyung Im Jung
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan, Republic of Korea
| | - Sang-Jae Lee
- Department of Bioscience and Research Center for Extremophiles and Marine Microbiology, Silla University, Busan, Republic of Korea
| | - Mi Hwa Park
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan, Republic of Korea
| |
Collapse
|
7
|
Yang B, Tan M, Xiong F. Global trends in osteoimmunology and osteoporosis research: A bibliometric analysis from 2013 to 2022. Medicine (Baltimore) 2025; 104:e42367. [PMID: 40324222 PMCID: PMC12055078 DOI: 10.1097/md.0000000000042367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/28/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND A large number of studies have shown that osteoporosis is closely related to bone immunology. The purpose of this study is to conduct bibliometrics and visual analysis of the fields related to osteoimmunology and osteoporosis from 2013 to 2022 and to summarize the research hotspots and trends in this field. METHODS We searched the Web of Science core collection database for articles on osteoimmunology and osteoporosis published between 2013 and 2022. Vosviewer 1.6.18 and CiteSpace.6.2. R4 were used to analyze the retrieved data. RESULTS A total of 3218 articles on osteoimmunology and osteoporosis were included in this study. A total of 76 countries, 347 institutions, and 502 authors were included in the articles examined in this study. The main research countries were China, the United States, and South Korea. Shanghai Jiaotong University, Harvard University, and the University of California system were the main research institutions. The author who published the most papers was Xu, Jiake. CONCLUSIONS This study is the first to summarize the global research trends in the field of osteoimmunology and osteoporosis from 2013 to 2022. That helps researchers quickly understand the research hotspots and directions in this field.
Collapse
Affiliation(s)
- Bencheng Yang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingshuai Tan
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fusheng Xiong
- Department of Spine Surgery, Suining Municipal Hospital of Traditional Chinese Medicine, Suining, Sichuan, China
| |
Collapse
|
8
|
Moharana SS, Mallick S, Pinto JR, Shenoy P S, Bose B. Osteo-inductive potential of homoeopathy potencies of Asafoetida on multipotent mesenchymal stem cells in vitro. 3 Biotech 2025; 15:124. [PMID: 40230371 PMCID: PMC11993510 DOI: 10.1007/s13205-025-04245-1] [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: 08/21/2024] [Accepted: 02/15/2025] [Indexed: 04/16/2025] Open
Abstract
The musculoskeletal system is the main framework of the human system. Poor osteogenesis is related to poor diet, improper exercise, autoimmune diseases, or age-related conditions. This tends to compromise the quality of human life, especially gait and mobility. Calcium, magnesium, and vitamin D supplements are the prescriptions that address the issues above. Moreover, various natural products in alternative medicines have also been found to have applications in improving bone health. One such finding in our study is the homeopathy potency of Asafoetida, 200CH, which has exhibited a promising osteo-inductive potential in vitro for murine multipotent mesenchymal stem cell line C3H10T1/2. Ferula asafoetida is otherwise a known herb for addressing gastric issues. In this work, the homoeopathy clinician (first author) first repurposed the F. asafoetida potency (dilution) to ameliorate bone disorders such as avascular necrosis and loose body dissolution. Following clinical success, we have proven the osteo-inductive properties of this particular homoeopathy potency of the drug in vitro. Most importantly, the osteo-inductive (osteogenesis) properties of homoeopathy potency 200CH are more pronounced than the native compound combinations in the Mother tincture (Q) of the drug. This homoeopathy drug, hence, can be used for treating osteoarthritis alone or else as an adjuvant with conventional treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04245-1.
Collapse
Affiliation(s)
- Sudhansu Sekhar Moharana
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka 575018 India
- Department of Homeopathic Repertory and Case Taking, Yenepoya Homeopathic Medical College and Hospital, Yenepoya (Deemed to Be University) Ayush Campus, Naringana, Deralakatte, Mangalore, Karnataka 575018 India
- Repertory Department, AGM Homeopathic Medical College and Hospital, Varur, Karnataka 581207 India
| | - Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka 575018 India
| | - Joel Rimson Pinto
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka 575018 India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka 575018 India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka 575018 India
| |
Collapse
|
9
|
Dong Y, Sun Y, Zhou Z, Gai Z, Cai Y, Han M, Zou K. Modulation of the gut-bone axis: Lacticaseibacillus paracasei LC86 improves bone health via anti-inflammatory metabolic pathways in zebrafish models of osteoporosis and cartilage damage. Front Immunol 2025; 16:1493560. [PMID: 40308595 PMCID: PMC12041650 DOI: 10.3389/fimmu.2025.1493560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Aim Osteoporosis and cartilage injury are major health concerns with limited treatment options. This study investigates the therapeutic effects of Lacticaseibacillus paracasei LC86 (LC86) on osteoporosis and cartilage damage in a zebrafish (Danio rerio) model, focusing on its modulation of the gut-bone axis and its potential mechanisms for enhancing bone health. Methods A Dexamethasone-induced zebrafish model was used to mimic osteoporosis and cartilage injury. Zebrafish were divided into control, model, and LC86 treatment groups (3×107 CFU/mL). Bone and cartilage health were assessed using Alizarin red staining and fluorescence microscopy. Bone marker expression (sp7, runx2a, bmp2a, bmp4, and col2a1a) was quantified via qPCR. Metabolic alterations were analyzed using untargeted metabolomics, and changes in gut microbiota were examined through 16S rRNA gene sequencing. Results LC86 treatment significantly improved bone and cartilage health, as evidenced by increased fluorescence intensity in the skull, hard bone, and cartilage (p < 0.01, p < 0.05). qPCR results showed upregulation of key bone-related genes (sp7, runx2a, bmp2a, bmp4, and col2a1a), indicating enhanced bone and cartilage structure. Metabolomics analysis revealed alterations in over 300 metabolites, with changes in anti-inflammatory and energy pathways. Gut microbiota analysis demonstrated an increase in beneficial bacteria and a decrease in pathogenic genera. Conclusions LC86 significantly improved bone health, cartilage structure, and gut microbiota composition in a Dexamethasone-induced zebrafish model, supporting its potential as a therapeutic strategy for osteoporosis and cartilage injury via modulation of the gut-bone axis.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Yukun Sun
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Zhipeng Zhou
- Food Science and Nutrition, University of Leeds, Leeds, United Kingdom
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Yihui Cai
- School of Biomedical Engineering, Hubei University of Medicine, Shiyan, China
| | - Mei Han
- Department of Food Quality and Safety, Shanghai Business School, Shanghai, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
10
|
Seo J, Ko R, Kim M, Seo J, Lee H, Kim D, Jeong W, Kim HS, Lee SY. Pim1 promotes the maintenance of bone homeostasis by regulating osteoclast function. Exp Mol Med 2025; 57:733-744. [PMID: 40164682 PMCID: PMC12046003 DOI: 10.1038/s12276-025-01421-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 04/02/2025] Open
Abstract
The Pim1 (proviral integration site for Moloney leukemia virus 1) protein is a serine/threonine kinase that is essential for cell proliferation, apoptosis and innate immune responses. Here we show that Pim1 promotes osteoclast resorptive function without affecting osteoclast numbers. Specifically, we found that mice lacking Pim1 (Pim1-/-) developed increased trabecular bone mass and indices such as trabecular bone-mass density. This was due to the direct phosphorylation of TRAF6 by Pim1 in mature osteoclasts, which activated the Akt-GSK3β signaling pathway. This, in turn, promoted the acetylation and consequent stabilization of microtubules, which permitted the formation of the osteoclast sealing zone. In vivo experiments then showed that, when mice with lipopolysaccharide-induced bone loss or tumor-induced osteolysis were treated with SGI-1776, a Pim1 inhibitor that is more selective for Pim1, the bone loss was significantly ameliorated. Thus, Pim1 plays an important role in osteoclast function and may be a therapeutic target for bone-related diseases.
Collapse
Affiliation(s)
- Jeongin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Ryeojin Ko
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Jeongmin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Woojin Jeong
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea.
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
11
|
Mamet T, Yang J, Zhang J, Guo Y, Zhao Z. Yak milk inhibits osteoclast differentiation by suppressing TRPV5 expression. J Dairy Sci 2025; 108:3142-3150. [PMID: 39824495 DOI: 10.3168/jds.2024-25607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/17/2024] [Indexed: 01/20/2025]
Abstract
Yak milk is a potential nutrient for improving osteoporosis. However, the effect of yak milk on the expression of Ca2+ion channel TRPV5 during osteoclast differentiation is still unclear. This study used ruthenium red as a control to investigate the effect of yak milk on osteoclast differentiation and activity. Tartrate-resistant acid phosphatase staining and bone resorption pit experiments showed that yak milk inhibited osteoclast differentiation and bone resorption activity in a dose-dependent manner. In addition, yak milk can inhibit osteoclast activity by inhibiting the expression of TRPV5. Quantitative real-time PCR and western blot results also exhibited that yak milk significantly decreased the expression of TRPV5 and calbindin-D28k mRNA and protein in osteoclasts. These results suggest that yak milk inhibits nuclear factor-κβ ligand-receptor activator-induced osteoclast differentiation and bone resorption activity in RAW 264.7 cells by suppressing the expression level of TRPV5 and calbindin-D28k mRNA and protein.
Collapse
Affiliation(s)
- Torkun Mamet
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830046, China.
| | - Jingru Yang
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jin Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yanping Guo
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Zhongkai Zhao
- Department of Food Science and Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830046, China
| |
Collapse
|
12
|
Li K, Cao H, Huang H, Tang S, Wang H, Yang Q, Hu Y, Weng J, Chen X. Advances in copper-containing biomaterials for managing bone-related diseases. Regen Biomater 2025; 12:rbaf014. [PMID: 40259976 PMCID: PMC12011366 DOI: 10.1093/rb/rbaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/10/2025] [Accepted: 03/08/2025] [Indexed: 04/23/2025] Open
Abstract
Bone-related diseases pose a major challenge in contemporary society, with significant implications for both health and economy. Copper, a vital trace metal in the human body, facilitates a wide range of physiological processes by being crucial for the function of proteins and enzymes. Numerous studies have validated copper's role in bone regeneration and protection, particularly in the development and expansion of bone collagen. Owing to copper's numerous biological advantages, an increasing number of scientists are endeavoring to fabricate novel, multifunctional copper-containing biomaterials as an effective treatment strategy for bone disorders. This review integrates the current understanding regarding the biological functions of copper from the molecular and cellular levels, highlighting its potential for bone regeneration and protection. It also reviews the novel fabrication techniques for developing copper-containing biomaterials, including copper-modified metals, calcium phosphate bioceramics, bioactive glasses, bone cements, hydrogels and biocomposites. The fabrication strategies and various applications of these biomaterials in addressing conditions such as fractures, bone tumors, osteomyelitis, osteoporosis, osteoarthritis and osteonecrosis are carefully elaborated. Moreover, the long-term safety and toxicity assessments of these biomaterials are also presented. Finally, the review addresses current challenges and future prospects, in particular the regulatory challenges and safety issues faced in clinical implementation, with the aim of guiding the strategic design of multifunctional copper-based biomaterials to effectively manage bone-related diseases.
Collapse
Affiliation(s)
- Kunwei Li
- School of Life Science and Engineering, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Huan Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Hao Huang
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Songyuan Tang
- School of Life Science and Engineering, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Han Wang
- Department of Cardiology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiao Tong University, Chengdu 610014, China
| | - Qing Yang
- Department of Cardiology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiao Tong University, Chengdu 610014, China
| | - Yonghe Hu
- College of Medicine, Southwest Jiao Tong University, Chengdu 610031, China
| | - Jie Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiao Tong University, Chengdu, Sichuan 610031, China
| | - Xin Chen
- School of Life Science and Engineering, Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| |
Collapse
|
13
|
Imamura K, Tachi K, Takayama T, Kasai H, Shohara R, Inoue K, Taguchi Y, Nakane-Koyachi S, Saito A, Yamano S. Developmental Endothelial Locus-1 Promotes Osteoclast Differentiation and Activation. Int J Mol Sci 2025; 26:2673. [PMID: 40141315 PMCID: PMC11942430 DOI: 10.3390/ijms26062673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Developmental endothelial locus-1 (DEL-1) has traditionally been characterized within the scientific community as having anti-inflammatory properties with potential inhibitory effects on osteoclast formation. Our investigation challenges this paradigm by examining Del-1 expression in RAW264.7 cells and bone marrow-derived macrophages (BMMs) during osteoclastogenesis, as well as its functional impact on osteoclast development and activity. Our experimental findings revealed that Del-1 mRNA levels were markedly elevated in cells stimulated by the receptor activator of the nuclear factor κB ligand compared to unstimulated precursors. When cultured with varying concentrations of recombinant DEL-1, osteoclast differentiation increased in a dose-dependent manner. Furthermore, BMMs isolated from ovariectomized mice exhibited significantly higher Del-1 mRNA expression than those from control animals. To confirm DEL-1's role, we employed RNA interference techniques, demonstrating that DEL-1 silencing in RAW264.7 cells substantially reduced osteoclast formation. These results suggest that DEL-1 plays a previously unrecognized role in promoting osteoclastogenesis and may contribute to bone metabolism imbalances in conditions like osteoporosis, highlighting its complex role in skeletal homeostasis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Kentaro Imamura
- Department of Periodontology, Tokyo Dental College, Chiyoda-ku, Tokyo 101-0061, Japan; (K.I.); (S.N.-K.); (A.S.)
- Oral Health Science Center, Tokyo Dental College, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Keita Tachi
- Komazawa Parkside Dental Clinic, Setagaya-ku, Tokyo 154-0021, Japan;
| | - Tadahiro Takayama
- Department of Periodontology, Nihon University School of Dentistry, Chiyoda-ku, Tokyo 101-0062, Japan;
| | - Hironori Kasai
- Kasai Dental Clinic, Kitakyushu-shi 800-0226, Fukuoka, Japan;
| | | | - Kenji Inoue
- Department of Prosthodontics, New York University College of Dentistry, New York, NY 10010, USA;
| | - Yoichiro Taguchi
- Department of Operative Dentistry, Endodontology and Periodontology, Matsumoto Dental University, Shiojiri-shi 399-0704, Nagano, Japan;
| | - Saki Nakane-Koyachi
- Department of Periodontology, Tokyo Dental College, Chiyoda-ku, Tokyo 101-0061, Japan; (K.I.); (S.N.-K.); (A.S.)
| | - Atsushi Saito
- Department of Periodontology, Tokyo Dental College, Chiyoda-ku, Tokyo 101-0061, Japan; (K.I.); (S.N.-K.); (A.S.)
- Oral Health Science Center, Tokyo Dental College, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Seiichi Yamano
- Department of Prosthodontics, New York University College of Dentistry, New York, NY 10010, USA;
| |
Collapse
|
14
|
Jin X, Wang W, Liu H, Zhao J, Li P, Li A, Song Z. Enhanced Bone Targeting of Poly(l-glutamic acid)s through Cationic or Aromatic Substitution. Biomacromolecules 2025; 26:1913-1922. [PMID: 39977118 DOI: 10.1021/acs.biomac.4c01714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Poly(l-glutamic acid)s (PLGs) are promising bone-targeting ligands due to their high molecular weight and facile preparation. Nevertheless, the bone-targeting efficiency of PLGs is still relatively low, validating the necessity to further enhance targeting through structural optimization. Herein, we report the use of a heteropolypeptide strategy to improve the bone targeting of PLGs through the incorporation of another side-chain functionality for enhanced affinity with bone tissues. Specifically, the introduction of cationic amino or aromatic phenolic side-chain residues resulted in a ∼2.3-fold or ∼1.6-fold increase in the in vivo bone targeting, respectively. Cationic modification not only improved the affinity with bone minerals but also exhibited prolonged retention in the bone tissues for more than 60 days. This work highlights the use of a heteropolypeptide library to screen and optimize the performance of polypeptide materials, offering promising bone-targeting polymeric materials for the design of bone-related nanomedicine.
Collapse
Affiliation(s)
- Xiaoxiong Jin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Wanying Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Hui Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jing Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Pengfei Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Aoting Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ziyuan Song
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
15
|
Wu Q, Zhou J, Du D, Guo B, Wang H, Lv W. Mongolian medicine Sugemule-7 decoction prevents osteoporosis via Erk1/2 and p38 MAPK signaling pathways according to network pharmacology analysis. Int J Biol Macromol 2025; 292:139166. [PMID: 39743063 DOI: 10.1016/j.ijbiomac.2024.139166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
Osteoporosis (OP) is a significant global public health concern that requires the development of safe and effective drugs for prevention and treatment. Sugemule-7 (SGML-7) decoction, a traditional Mongolian herbal prescription, has long been used for treating OP, but its components and mechanisms of action remain unclear. The study identified the main compounds of SGML-7 using UHPLC-Q Exactive MS and explored the multi-target mechanisms of SGML-7 in OP through network pharmacology and molecular docking. A retinoic acid (RA)-induced mouse OP model was utilized to confirm the therapeutic effects and potential mechanism of SGML-7. Additionally, mouse pre-osteoblastic (MC3T3-E1) cells treated with SGML-7 medicated serum were employed to delve deeper into the molecular mechanisms. The UHPLC-Q Exactive MS analysis, network pharmacology, and molecular docking suggested that the synergistic effect of multiple active compounds could be the main contributor to SGML-7 for its anti-OP activities. Moreover, MAPK1, JUN, ESR1, TP53, AKT1, NCOA1, FOS, and NR3C1 were identified as potential key targets, and the MAPK signaling pathway was among the signaling pathways possibly involved in the anti-OP activities of SGML-7. Consistent with these findings, experimental studies confirmed that SGML-7 prevented bone loss, enhanced bone quality in OP mice, and promoted osteoblastic activity and bone formation in MC3T3-E1 cells by modulating MAPK-associated targets. Taken together, SGML-7 shows promise as an effective and appealing anti-OP drug candidate.
Collapse
Affiliation(s)
- Qijin Wu
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| | - Jing Zhou
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Donghua Du
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Bing Guo
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Haifeng Wang
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Wenting Lv
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China.
| |
Collapse
|
16
|
Kim I, Park S, Kim J, Park SY, Seo J, Roh S. Treatment with Lactobacillus paracasei L30 extract induces osteogenic differentiation of human bone marrow mesenchymal stem cells in vitro. Biomed Pharmacother 2025; 184:117913. [PMID: 39955853 DOI: 10.1016/j.biopha.2025.117913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/22/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025] Open
Abstract
Bone-related diseases such as osteoporosis pose a significant health economic burden to countries around the world and, because current treatments are insufficient, more effective therapies are desperately needed. This study explored the potential of Lactobacillus paracasei L30 extract to influence the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs). Our results showed that L30 extract significantly enhanced the expression of osteogenic markers in hBM-MSCs, including alkaline phosphatase (ALP), runt-related transcription factor 2 (RUNX2), and collagen type I alpha 1 (COL1A1). Mechanistic studies revealed that L30 extract activated the p38 MAPK and AKT signaling pathways, leading to phosphorylation of Glycogen synthase kinase-3 beta (GSK3β) and subsequent nuclear translocation of β-catenin. Conversely, inhibition of p38 MAPK, AKT, or knockdown of β-catenin significantly attenuated the osteogenic effects of L30 extract on hBM-MSCs. Furthermore, we found that L30 extract promoted osteogenic differentiation in primary osteoblast precursors isolated from mouse calvaria and enhances bone formation in ex vivo calvarial organ cultures. Therefore, the application of Lactobacillus paracasei L30 extract in such contexts could serve as a therapeutic approach for promoting bone formation. Collectively, our findings suggest a novel approach for the clinical management of bone-related disorders, with possible applications for treating diseases such as osteoporosis.
Collapse
Affiliation(s)
- Inwook Kim
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seocho-gu 06663, Republic of Korea
| | - Sankyu Park
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seocho-gu 06663, Republic of Korea
| | - Jieun Kim
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seocho-gu 06663, Republic of Korea
| | - So Young Park
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seocho-gu 06663, Republic of Korea
| | - Jeongmin Seo
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Seocho-gu 06663, Republic of Korea; Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Republic of Korea.
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Republic of Korea.
| |
Collapse
|
17
|
Xu H, Zhou Z, Wen F, Sun H, Hou J. Inhibiting autophagy further promotes Ginkgolide B's anti-osteoclastogenesis ability. Bone 2025; 192:117348. [PMID: 39617111 DOI: 10.1016/j.bone.2024.117348] [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: 09/08/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Excessive osteoclast activity could cause skeletal diseases including osteoporosis. Additionally, autophagy plays an initial role in osteoclast differentiation and function. Ginkgolide B (GB), a key compound in Ginkgo biloba, improves bone mass and suppresses mature osteoclast formation in vitro. This study examines the role of GB role in regulating osteoclast formation via autophagy. Using murine bone marrow-derived macrophages in vitro, we explored GB's effects on autophagy and osteoclast formation. We also assessed bone loss prevention in an ovariectomized (OVX) mouse model using GB combined with an autophagy inhibitor. Tartrate-resistant acid phosphatase staining was used to observe osteoclast formation. Autophagy-related proteins and intracellular microtubule associated protein 1 light chain 3 beta puncta were observed using western blotting and immunofluorescence. The impact of GB on OVX mice was evaluated using micro-computed tomography and hematoxylin and eosin staining. GB directly promoted autophagy in osteoclast precursors (OCPs), but inhibited osteoclast differentiation by reducing receptor activator of nuclear factor kappa B ligand (RANKL)-induced autophagy. GB inhibits the phosphorylation of RANKL downstream signaling pathways, such as Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK). Anisomycin (ANI), a JNK activator, reversed GB's inhibitory effects on RANKL-induced autophagy and osteoclastogenesis. Inhibiting autophagy using 3-Methyladenine (3-MA) or small interfering RNA significantly suppressed osteoclast differentiation both in vitro and in vivo. Our findings suggested that GB inhibits osteoclast formation by decreasing JNK phosphorylation and autophagy under RANKL stimulation. Interestingly, GB also directly promotes autophagy in OCPs. Thus, GB markedly reduces osteoclast differentiation and prevents bone loss, with its anti-osteoclastogenesis effect being enhanced by 3-MA. Accordingly, inhibiting GB-induced direct autophagy could further increase its therapeutic effect on bone disease resulting from excessive osteoclast activity.
Collapse
Affiliation(s)
- Haoying Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hematology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Zijie Zhou
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Orthopedics, Orthopedic and Sports Medicine Center, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China
| | - Fuli Wen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China
| | - Hong Sun
- Department of Pharmacy, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China.
| | - Jianming Hou
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, China.
| |
Collapse
|
18
|
Sharan K, Brandt C, Yusuf MA, Singh P, Halder N, Edwards ME, Mangu SVVSR, Das A, Mishra A, Kumar SS, Sharma A, Gupta A, Liu XS, Guo EX, Monani UR, Ponnalagu D, Ivanov II, Lal G, Clare S, Dougan G, Yadav VK. Rapid and relaying deleterious effects of a gastrointestinal pathogen, Citrobacter rodentium, on bone, an extra-intestinal organ. iScience 2025; 28:111802. [PMID: 39967874 PMCID: PMC11834125 DOI: 10.1016/j.isci.2025.111802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/04/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Enteropathogenic infections cause pathophysiological changes in the host but their effects beyond the gastrointestinal tract are undefined. Here, using Citrobacter rodentium infection in mouse, which mimics human diarrheal enteropathogenic Escherichia coli, we show that gastrointestinal infection negatively affects bone remodeling, leading to compromised bone architecture. Transmission of infection through fecal-oral route from Citrobacter rodentium-infected to non-infected mice caused bone loss in non-infected cage mates. Mice with B cell deficiency (Igh6-/- mice) failed to clear C. rodentium infection and exhibited more severe and long-term bone loss compared to WT mice. Unbiased cytokine profiling showed an increase in circulating tumor necrosis factor α (TNFα) levels following Citrobacter rodentium infection, and immunoneutralization of TNFα prevented infection-induced bone loss completely in WT and immunocompromised mice. These findings reveal rapid, relaying, and modifiable effects of enteropathogenic infections on an extraintestinal organ-bone, and provide insights into the mechanism(s) through which these infections affect extraintestinal organ homeostasis.
Collapse
Affiliation(s)
- Kunal Sharan
- Mouse Genetics Project, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Cordelia Brandt
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
| | - Mohd Aslam Yusuf
- Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Parminder Singh
- National Institute of Immunology, New Delhi, New Delhi, India
| | - Namrita Halder
- National Centre for Cell Science, Pune, Maharastra, India
| | - Madeline E. Edwards
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - SVVS Ravi Mangu
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Abhilipsa Das
- Department of Molecular Nutrition, CSIR-CFTRI, Mysore, Karnataka, India
| | - Amrita Mishra
- National Centre for Cell Science, Pune, Maharastra, India
| | - Shashi S. Kumar
- Center for Motor Neuron Biology & Disease, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Amita Sharma
- Pediatric Kidney Foundation, New Delhi, New Delhi, India
| | - Alka Gupta
- Reproductive Biology Laboratory, National Institute of Immunology, New Delhi, New Delhi, India
| | - Xiaowei S. Liu
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward X. Guo
- Bone Biomechanics Laboratory, Columbia University, New York, NY, USA
| | - Umrao R. Monani
- Center for Motor Neuron Biology & Disease, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | | | - Ivaylo I. Ivanov
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Girdhari Lal
- National Centre for Cell Science, Pune, Maharastra, India
| | - Simon Clare
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
| | - Gordon Dougan
- Host-Pathogen Interaction Group, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Centre for Translational Stem Cell Biology, Hong Kong, China
| | - Vijay K. Yadav
- Mouse Genetics Project, Wellcome Sanger Institute, Hinxton, Saffron Walden, UK
- National Institute of Immunology, New Delhi, New Delhi, India
- Department of Genetics and Development, Columbia University, New York, NY, USA
- Healthy Longevity Program, Department of Pathology, Immunology and Laboratory Medicine, Rutgers University, Newark, NJ, USA
- Center for Cell Signaling, Rutgers University, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers University, Newark, NJ, USA
| |
Collapse
|
19
|
Vijayakumar S, González-Sánchez ZI, Amanullah M, Sonamuthu J, Rajkumar M, Divya M, Durán-Lara EF, Li M. Shark chondroitin sulfate gold nanoparticles: A biocompatible apoptotic agent for osteosarcoma. Int J Biol Macromol 2025; 290:138793. [PMID: 39689798 DOI: 10.1016/j.ijbiomac.2024.138793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Osteosarcoma is a highly aggressive tumor that originates in the bone and often infiltrates nearby bone cells. It is the most prevalent type of primary bone cancer among the various bone malignancies. Traditional cancer treatment methods such as surgery, chemotherapy, immunotherapy, and radiotherapy have had restricted success. However, the integration of nanotechnology into cancer research has led to notable progress. One promising area is the use of marine-derived polysaccharide-based nano formulations for treating various human diseases, including cancer. This study presents a straightforward method for synthesizing biocompatible gold nanoparticles (AuNPs), utilizing sodium borohydride as a reducing agent and a cost-effective, water-soluble chondroitin sulfate (CS) derived from shark cartilage as a stabilizing agent. The synthesized CS-Au NPs appeared purple and were mainly spherical, with 40.768 nm of average size. Cytotoxicity assays (MTT) indicated that CS-Au NPs significantly reduced the viability of human osteosarcoma cells (MG63) at 100 μg/mL, while it showed no cytotoxic effects on mouse embryonic fibroblast cells (NIH3T3) at the same concentration. The observed toxicity of the CS-Au NPs was linked to a rise in the production of reactive oxygen species (ROS) within damaged mitochondria. ROS generation and changes in mitochondrial membrane potential were detected in MG63 cells treated with CS-Au NPs. Furthermore, apoptotic analysis through ethidium bromide dual staining and flow cytometry demonstrated that CS-Au NPs at higher concentrations significantly increased the amount of apoptotic cells, as demonstrated by acridine orange/ethidium bromide staining. Flow cytometry also confirmed that CS-Au NPs activated the apoptotic pathway in MG63 cells.
Collapse
Affiliation(s)
- Sekar Vijayakumar
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| | - Zaira I González-Sánchez
- Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, PUCMM, Autopista Duarte Km 1 ½, Santiago de los Caballeros, Dominican Republic; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Jegatheeswaran Sonamuthu
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mangaiyarkarasi Rajkumar
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mani Divya
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Esteban F Durán-Lara
- Bio&NanoMaterialsLab Drug Delivery and Controlled Release, Universidad de Talca, Talca 3460000, Maule, Chile; Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Maule, Chile
| | - Mingchun Li
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| |
Collapse
|
20
|
Yoon I, Xue L, Chen Q, Liu J, Xu J, Siddiqui Z, Kim D, Chen B, Shi Q, Laura Han E, Cherry Ruiz M, Vining KH, Mitchell MJ. Piperazine-Derived Bisphosphonate-Based Ionizable Lipid Nanoparticles Enhance mRNA Delivery to the Bone Microenvironment. Angew Chem Int Ed Engl 2025; 64:e202415389. [PMID: 39379320 PMCID: PMC11735871 DOI: 10.1002/anie.202415389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
Nucleic acid delivery with mRNA lipid nanoparticles are being developed for targeting a wide array of tissues and cell types. However, targeted delivery to the bone microenvironment remains a significant challenge in the field, due in part to low local blood flow and poor interactions between drug carriers and bone material. Here we report bone-targeting ionizable lipids incorporating a piperazine backbone and bisphosphate moieties, which bind tightly with hydroxyapatite ([Ca5(PO4)3OH]), a key component of mineralized tissues. These lipids demonstrate biocompatibility and low toxicity in both vitro and in vivo studies. LNP formulated with these lipids facilitated efficient cellular transfection and improved binding to hydroxyapatite in vitro, and targeted delivery to the bone microenvironment in vivo following systemic administration. Overall, our findings demonstrate the critical role of the piperazine backbone in a novel ionizable lipid, which incorporates a bisphosphonate group to enable efficient bone-targeted delivery, highlighting the potential of rational design of ionizable lipids for next-generation bone-targeting delivery systems.
Collapse
Affiliation(s)
- Il‐Chul Yoon
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Department of Materials Science and EngineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Lulu Xue
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Qinyuan Chen
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Jingyi Liu
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Junchao Xu
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Zain Siddiqui
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Dongyoon Kim
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Bingling Chen
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Qiangqiang Shi
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Emily Laura Han
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Mia Cherry Ruiz
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Kyle H. Vining
- Department of Materials Science and EngineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Preventive and Restorative SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| | - Michael J. Mitchell
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Abramson Cancer CenterPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Center for Cellular ImmunotherapiesPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Penn Institute for RNA InnovationPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Institute for ImmunologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Cardiovascular InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
- Institute for Regenerative MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104United States
| |
Collapse
|
21
|
Tong X, Fu X, Gong A, Yu G, Chen N, Chen B, Gu J, Liu Z. Effect of Luteolin on cadmium-inhibited bone growth via suppressing osteoclastogenesis in laying chickens. J Anim Sci 2025; 103:skaf033. [PMID: 39921628 PMCID: PMC11912829 DOI: 10.1093/jas/skaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/07/2025] [Indexed: 02/10/2025] Open
Abstract
Luteolin (Lut) is a flavonoid derived from several plant sources. Cadmium (Cd) is a widespread environmental contaminant and potential toxin with detrimental effects on animal health. However, the effect of Lut on Cd-induced inhibition of bone growth in laying chickens remains unclear. This study investigates the effects of Lut on Cd-induced inhibition of bone growth in the femur and tibia of laying chickens. A total of sixty 1-d-old green-eggshell yellow feather laying chickens were randomly assigned to 4 groups after a 5-d acclimation period: basal diet (Con), cadmium chloride (CdCl2, Cd), Lut, and Lut + Cd. Bone microstructure, serum biomarkers of bone remodeling, the levels of Cd, calcium (Ca), phosphorus (P), and trace metal elements were assessed using the micro-computed tomography (Micro-CT), enzyme-linked immunosorbent assay (ELISA), and microwave digestion, respectively. Bone remodeling biomarkers, late endosomal/lysosomal adaptor and MAPK and mTOR activator 1 (LAMTOR1), as well as the phosphorylation of AMP-activated protein kinase α (AMPKα) and protein kinase B (Akt), were quantified using the qRT-PCR and western blot. The results indicated that Lut effectively mitigated Cd-induced bone mass loss compared to the Cd group, resulting in increased bone volume (BV), bone surface/BV (BS/BV), connectivity density (Conn.Dn), and the length and weight of the femur and tibia in laying chickens. Mechanistically, compared to the Cd group, Lut restored the ratio of osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) in serum and bone tissue, enhanced the expression of bone morphogenetic protein-2 (BMP-2), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and Osterix (OSX), while reducing the levels of Ca, Cd, and alkaline phosphatase (ALP) activity, as well as the expression of osteopontin (OPN), c-Fos, osteoclast stimulatory-transmembrane protein (OC-STAMP), tartrate-resistant acid phosphatase, cathepsin K (CTSK), matrix metalloprotein-9 (MMP-9), LAMTOR1, and the phosphorylation of AMPKα and Akt. Therefore, Lut alleviates Cd-induced damage to the femur and tibia of chickens by promoting osteogenesis and inhibiting osteoclastogenesis, positioning Lut as a potential therapeutic plant extract for enhancing bone growth in laying chickens.
Collapse
Affiliation(s)
- Xishuai Tong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Xiaohui Fu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Anqing Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Gengsheng Yu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Naineng Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Bing Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Jianhong Gu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Zongping Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
22
|
Jalil AT, Abdulhadi MA, Al-Ameer LR, Abd-Alzahraa ZH, Merza MS, Zabibah RS, Bahair H, Yaas MH. Osteoporosis in Adrenal Insufficiency: Could Metformin be Protective? Indian J Clin Biochem 2025; 40:4-11. [PMID: 39835225 PMCID: PMC11741967 DOI: 10.1007/s12291-023-01153-0] [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: 06/26/2023] [Accepted: 09/01/2023] [Indexed: 01/22/2025]
Abstract
Adrenal insufficiency (AI) is a serious disorder characterized by the adrenal glucocorticoid deficiency. Regardless of the etiology, AI patients need long-term replacement therapy for glucocorticoids and, in some cases, for mineralocorticoids. The replacement therapy cannot completely mirror the physiological secretion patterns, and therefore, glucocorticoid excess is a common sequela in AI patients. Moreover, due to the absence of the reliable clinical markers to monitor the adequacy of the replacement therapy, clinicians often over-treat the AI patients to avoid adrenal crisis. Long-term glucocorticoid use is associated with the loss of bone density and osteoporosis, increasing the risk of fractures. Moreover, glucocorticoid-induced hyperglycemia and type 2 diabetes mellitus further aggravates the bone disorders. In the recent years, ameliorating effects of metformin on glucocorticoid-induced bone disorders, as well as hyperglycemia, have been reported by a multitude of studies; and here, we reviewed and discussed the most recent findings regarding the positive effects of metformin on alleviating the bone disorders, and their implications in the AI patients.
Collapse
Affiliation(s)
| | - Mohanad Ali Abdulhadi
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | | | - Muna S. Merza
- Prosthetic Dental Techniques Department, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Rahman S. Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University of Najaf, Najaf, Iraq
| | - Hala Bahair
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
23
|
Sakai E, Tsukuba T. Transcriptomic Characterization Reveals Mitochondrial Involvement in Nrf2/Keap1-Mediated Osteoclastogenesis. Antioxidants (Basel) 2024; 13:1575. [PMID: 39765903 PMCID: PMC11673794 DOI: 10.3390/antiox13121575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress regulation in osteoclastogenesis, DNA microarray analysis was conducted in this study using wild-type (WT), Keap1 knockout (Keap1 KO), and Nrf2 knockout (Nrf2 KO) osteoclasts. Principal component analysis showed that 403 genes, including Nqo1, Il1f9, and Mmp12, were upregulated in Keap1 KO compared with WT osteoclasts, whereas 24 genes, including Snhg6, Ccdc109b, and Wfdc17, were upregulated in Nrf2 KO compared with WT osteoclasts. Moreover, 683 genes, including Car2, Calcr, and Pate4, were upregulated in Nrf2 KO cells compared to Keap1 KO cells. Functional analysis by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed upregulated genes in Nrf2 KO osteoclasts were mostly enriched in oxidative phosphorylation. Furthermore, GeneMANIA predicted the protein-protein interaction network of novel molecules such as Rufy4 from genes upregulated in Nrf2 KO osteoclasts. Understanding the complex interactions between these molecules may pave the way for developing promising therapeutic strategies against bone metabolic diseases caused by increased osteoclast differentiation under oxidative stress.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan;
| | | |
Collapse
|
24
|
Qi ML, Li M, Yuan K, Song E, Zhang H, Yao S. Fabrication and X-ray microtomography of sandwich-structured PEEK implants for skull defect repair. Sci Rep 2024; 14:28585. [PMID: 39562669 PMCID: PMC11577077 DOI: 10.1038/s41598-024-80103-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024] Open
Abstract
Bone defects pose a significant risk to human health. Medical polyetheretherketone (PEEK) is an excellent implant material for bone defect repair, but it faces the challenge of bone osteoconduction and osseointegration. Osteoconduction describes the process by which bone grows on the surface of the implant, while osseointegration is the stable anchoring of the implant achieved by direct contact between the bone and the implant. Bone defects repair depends on the implant's three-dimensional spatial structure, including pore size, porosity, and interconnections to a great extent. However, it is challenging to fabricate the porous structures to meet specific requirements and to characterize them without causing damage. In this study, we designed and fabricated sandwich-like PEEK implants mimicking the three-layer structures of the skull, whose defects imposes a significant burden on young adulthood and paediatric populations, and performed in-line phase-contrast synchrotron X-ray microtomography to non-destructively investigate the internal porous microstructures. The sandwich-like three-layer microstructure, comprising a dense layer, a loose layer and a dense layer in succession, exhibits structural similarity to that in a natural skull. This work demonstrated the fabrication of the sandwich-like PEEK implant that could potentially enhance osteoconduction and osseointegration. Furthermore, the interior structures and residual porogen sodium chloride particles were observed within the PEEK implant, which cannot be realized by other microscopic methods without destroying the sample. It highlights the advantages and potential of using synchrotron X-ray microtomography to analyze the structure of biomedical materials. This study provides theoretical guidance for the further design and fabrication of PEEK bone repair materials and will advance the clinical application of innovative bioactive bone repair materials.
Collapse
Affiliation(s)
- Mei-Li Qi
- School of Civil Engineering, Shandong Jiaotong University, Jinan, 250357, China
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Minghua Li
- School of Civil Engineering, Shandong Jiaotong University, Jinan, 250357, China
| | - Kunshan Yuan
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Enhui Song
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China
| | - Haijun Zhang
- National United Engineering Laboratory for Biomedical Material Modification, Dezhou, 251100, China.
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Shengkun Yao
- Shandong Provincial Engineering and Technical Center of Light Manipulations & Shandong Provincial Key Laboratory of Optics and Photonic Device, School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
- Collaborative Innovation Center of Light Manipulation and Applications, Shandong Normal University, Jinan, 250358, China.
| |
Collapse
|
25
|
Li W, Li L, Hu J, Zhou D, Su H. Design and Applications of Supramolecular Peptide Hydrogel as Artificial Extracellular Matrix. Biomacromolecules 2024; 25:6967-6986. [PMID: 39418328 DOI: 10.1021/acs.biomac.4c00971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Supramolecular peptide hydrogels (SPHs) consist of peptides containing hydrogelators and functional epitopes, which can first self-assemble into nanofibers and then physically entangle together to form dynamic three-dimensional networks. Their porous structures, excellent bioactivity, and high dynamicity, similar to an extracellular matrix (ECM), have great potential in artificial ECM. The properties of the hydrogel are largely dependent on peptides. The noncovalent interactions among hydrogelators drive the formation of assemblies and further transition into hydrogels, while bioactive epitopes modulate cell-cell and cell-ECM interactions. Therefore, SPHs can support cell growth, making them ideal biomaterials for ECM mimics. This Review outlines the classical molecular design of SPHs from hydrogelators to functional epitopes and summarizes the recent advancements of SPHs as artificial ECMs in nervous system repair, wound healing, bone and cartilage regeneration, and organoid culture. This emerging SPH platform could provide an alternative strategy for developing more effective biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Wenting Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Longjie Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jiale Hu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Dongdong Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
26
|
Yao M, Li M, Peng D, Wang Y, Li S, Zhang D, Yang B, Qiu HJ, Li LF. Unraveling Macrophage Polarization: Functions, Mechanisms, and "Double-Edged Sword" Roles in Host Antiviral Immune Responses. Int J Mol Sci 2024; 25:12078. [PMID: 39596148 PMCID: PMC11593441 DOI: 10.3390/ijms252212078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Numerous viruses that propagate through the respiratory tract may be initially engulfed by macrophages (Mφs) within the alveoli, where they complete their first replication cycle and subsequently infect the adjacent epithelial cells. This process can lead to significant pathological damage to tissues and organs, leading to various diseases. As essential components in host antiviral immune systems, Mφs can be polarized into pro-inflammatory M1 Mφs or anti-inflammatory M2 Mφs, a process involving multiple signaling pathways and molecular mechanisms that yield diverse phenotypic and functional features in response to various stimuli. In general, when infected by a virus, M1 macrophages secrete pro-inflammatory cytokines to play an antiviral role, while M2 macrophages play an anti-inflammatory role to promote the replication of the virus. However, recent studies have shown that some viruses may exhibit the opposite trend. Viruses have evolved various strategies to disrupt Mφ polarization for efficient replication and transmission. Notably, various factors, such as mechanical softness, the altered pH value of the endolysosomal system, and the homeostasis between M1/M2 Mφs populations, contribute to crucial events in the viral replication cycle. Here, we summarize the regulation of Mφ polarization, virus-induced alterations in Mφ polarization, and the antiviral mechanisms associated with these changes. Collectively, this review provides insights into recent advances regarding Mφ polarization in host antiviral immune responses, which will contribute to the development of precise prevention strategies as well as management approaches to disease incidence and transmission.
Collapse
Affiliation(s)
- Meng Yao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Dingkun Peng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Yijing Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (M.Y.); (M.L.); (D.P.); (Y.W.); (S.L.)
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China; (D.Z.); (B.Y.)
| |
Collapse
|
27
|
Gal M, Tuan HM, Park JH, Park KH, Kim O, Min BS, Lee JH. Irilin D suppresses RANKL-induced osteoclastogenesis and prevents inflammation-induced bone loss by disrupting the NF-κB and MAPK signaling pathways. Eur J Pharmacol 2024; 982:176956. [PMID: 39209096 DOI: 10.1016/j.ejphar.2024.176956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Excessive activity of osteoclasts(OCs) lead to bone resorption in chronic inflammatory conditions. The use of natural compounds to target OCs offers significant promise in the treatment or prevention of OC-associated diseases. Irilin D (IRD), a natural isoflavone derived from Belamcanda chinensis (L.) DC., has potential effects on OC differentiation both in vitro and in vivo that have yet to be thoroughly explored. In our study, we found that IRD inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced OC differentiation, actin ring formation, and bone resorption in vitro without compromising cell viability. However, IRD did not exhibit anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated macrophages. Furthermore, IRD reduced LPS-induced inflammatory bone loss by blocking osteoclastogenesis in a mouse model. Mechanistically, IRD disrupted RANKL-induced activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB), leading to the inhibition of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) activation. We also demonstrated that IRD inhibited RANKL-induced osteoclastic NFATc1 target genes, including DC-STAMP, ACP5, and CtsK. Our results indicate that IRD mitigates LPS-induced inflammatory bone resorption in mice by inhibiting RANKL-activated MAPKs and NF-κB signaling pathways, suggesting its potential as a natural isoflavone for preventing or treating OC-associated diseases.
Collapse
Affiliation(s)
- Minju Gal
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Ha Manh Tuan
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk, 38430, Republic of Korea
| | - Ju-Hee Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Kang-Hyeon Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Okhwa Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Byung-Sun Min
- College of Pharmacy, Drug Research and Development Center, Daegu Catholic University, Gyeongbuk, 38430, Republic of Korea.
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea.
| |
Collapse
|
28
|
Lin Y, Chen M, Guo W, Qiu S, Chen L, Liu W. Zoledronic acid relieves steroid-induced avascular necrosis of femoral head via inhibiting FOXD3 mediated ANXA2 transcriptional activation. Bone 2024; 188:117222. [PMID: 39102974 DOI: 10.1016/j.bone.2024.117222] [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: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Zoledronic acid (ZOL) is a type of bisphosphonate with good therapeutic effects on orthopaedic diseases. However, the pharmacological functions of ZOL on steroid-induced avascular necrosis of femoral head (SANFH) and the underlying mechanism remain unclear, which deserve further research. METHODS SANFH models both in vivo and in vitro were established by dexamethasone (Dex) stimulation. Osteoclastogenesis was examined by TRAP staining. Immunofluorescence was employed to examine autophagy marker (LC3) level. Cell apoptosis was analyzed by TUNEL staining. The interaction between Foxhead box D3 protein (FOXD3) and Annexin A2 (ANXA2) promoter was analyzed using ChIP and dual luciferase reporter gene assays. RESULTS Dex aggravated osteoclastogenesis and induced osteoclast differentiation and autophagy in vitro, which was abrogated by ZOL treatment. PI3K inhibitor LY294002 abolished the inhibitory effect of ZOL on Dex-induced osteoclast differentiation and autophagy. FOXD3 overexpression neutralized the downregulation effects of ZOL on Dex-induced osteoclasts by transcriptionally activating ANXA2. ANXA2 knockdown reversed the effect of FOXD3 overexpression on ZOL-mediated biological effects in Dex-treated osteoclasts. In addition, ZOL improved SANFH symptoms in rats. CONCLUSION ZOL alleviated SANFH through regulating FOXD3 mediated ANXA2 transcriptional activity and then promoting PI3K/AKT/mTOR pathway, revealing that FOXD3 might be a target for ZOL in SANFH treatment.
Collapse
Affiliation(s)
- Yu Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350000, Fujian Province, PR China; Department of Orthopedics, Fujian Pingtan Comprehensive Experimental Area Hospital, Fuzhou 350400, Fujian Province, PR China
| | - Min Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Wenbin Guo
- Department of Pathology, Pingtan Comprehensive Experimental Area Hospital, Fuzhou 350400, Fujian Province, PR China
| | - Shengliang Qiu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou 350000, Fujian Province, PR China
| | - Lihui Chen
- Laboratory Medicine, Fujian Pingtan Comprehensive Experimental Area Hospital, Fuzhou 350400, Fujian Province, PR China.
| | - Wenge Liu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou 350000, Fujian Province, PR China.
| |
Collapse
|
29
|
Chen Y, Chen QW, Fu FS, Gu HY, Yu A, Zhang XZ. Bone Destruction-Chemotactic Osteoprogenitor Cells Deliver Liposome Nanomedicines for the Treatment of Osteosarcoma and Osteoporosis. ACS NANO 2024; 18:29864-29879. [PMID: 39424791 DOI: 10.1021/acsnano.4c10053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Therapeutic efficacy of skeletal diseases is usually limited by unfavorable drug delivery due to incapable bone targeting and low bone affinity of conventional drug carriers, as well as relatively reduced vascularization and dense structure of bone tissues. Due to CXC chemokine receptor 4 (CXCR4)/CXC chemokine ligand 12 (CXCL12) signal axis-guided recruitment, osteoprogenitor cells (OPCs) can actively migrate to bone disease nidus. Here, drugs-loaded nanoliposomes are prepared and decorated onto OPCs by biotin-streptavidin linkage for precise bone disease targeting and effective drug delivery. In mouse models of tibia defect and orthotopic osteosarcoma, superior targeting property of OPCs-based drug delivery systems toward diseased bone niduses is verified. By encapsulating antitumor and antiosteoporosis drugs into nanoliposomes, OPCs-based drug delivery systems effectively inhibit disease development and restore bone destruction in mouse models of orthotopic osteosarcoma and ovariectomized osteoporosis. This study reveals a cell-based drug delivery system for precise bone disease targeting and highly effective drug delivery, which will find great potential as a universal drug delivery platform for targeting treatment of various bone diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Hui-Yun Gu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
30
|
Sakai E, Saito M, Koyanagi Y, Takayama Y, Farhana F, Yamaguchi Y, Tsukuba T. Autophagy Regulator Rufy 4 Promotes Osteoclastic Bone Resorption by Orchestrating Cytoskeletal Organization via Its RUN Domain. Cells 2024; 13:1766. [PMID: 39513873 PMCID: PMC11545195 DOI: 10.3390/cells13211766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Rufy4, a protein belonging to the RUN and FYVE domain-containing protein family, participates in various cellular processes such as autophagy and intracellular trafficking. However, its role in osteoclast-mediated bone resorption remains uncertain. In this study, we investigated the expression and role of the Rufy4 gene in osteoclasts using small interfering RNA (siRNA) transfection and gene overexpression systems. Our findings revealed a significant increase in Rufy4 expression during osteoclast differentiation. Silencing Rufy4 enhanced osteoclast differentiation, intracellular cathepsin K levels, and formation of axial protrusive structures but suppressed bone resorption. Conversely, overexpressing wild-type Rufy4 in osteoclasts hindered differentiation while promoting podosome formation and bone resorption. Similarly, overexpression of a Rufy4 variant lacking the RUN domain mimics the effects of Rufy4 knockdown, significantly increasing intracellular cathepsin K levels, promoting osteoclastogenesis, and elongated axial protrusions formation, yet inhibiting bone resorption. These findings indicate that Rufy4 plays a critical role in osteoclast differentiation and bone resorption by regulating the cytoskeletal organization through its RUN domain. Our study provides new insights into the molecular mechanisms governing osteoclast activity and underscores Rufy4's potential as a novel therapeutic target for bone disorders characterized by excessive bone resorption.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Minoru Saito
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Kondou Dental Clinic, 1154-5 Oozujinnai, Kikuchi 869-1221, Japan
| | - Yu Koyanagi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Department of Prosthetic Dentistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Yoshitsugu Takayama
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Ito Dental Clinic Medical Corporation, 3-2-4 Kousienn, Nishinomiya 663-8152, Japan
| | - Fatima Farhana
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Yu Yamaguchi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| |
Collapse
|
31
|
Moraiti S, Cheong VS, Dall’Ara E, Kadirkamanathan V, Bhattacharya P. A novel framework for elucidating the effect of mechanical loading on the geometry of ovariectomized mouse tibiae using principal component analysis. Front Bioeng Biotechnol 2024; 12:1469272. [PMID: 39502499 PMCID: PMC11534826 DOI: 10.3389/fbioe.2024.1469272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Murine models are used to test the effect of anti-osteoporosis treatments as they replicate some of the bone phenotypes observed in osteoporotic (OP) patients. The effect of disease and treatment is typically described as changes in bone geometry and microstructure over time. Conventional assessment of geometric changes relies on morphometric scalar parameters. However, being correlated with each other, these parameters do not describe separate fractions of variations and offer only a moderate insight into temporal changes. Methods The current study proposes a novel image-based framework that employs deformable image registration on in vivo longitudinal images of bones and Principal Component Analysis (PCA) for improved quantification of geometric effects of OP treatments. This PCA-based model and a novel post-processing of score changes provide orthogonal modes of shape variations temporally induced by a course of treatment (specifically in vivo mechanical loading). Results and Discussion Errors associated with the proposed framework are rigorously quantified and it is shown that the accuracy of deformable image registration in capturing the bone shapes (∼1 voxel = 10.4 μm) is of the same order of magnitude as the relevant state-of-the-art evaluation studies. Applying the framework to longitudinal image data from the midshaft section of ovariectomized mouse tibia, two mutually orthogonal mode shapes are reliably identified to be an effect of treatment. The mode shapes captured changes of the tibia geometry due to the treatment at the anterior crest (maximum of 0.103 mm) and across the tibia midshaft section and the posterior (0.030 mm) and medial (0.024 mm) aspects. These changes agree with those reported previously but are now described in a compact fashion, as a vector field of displacements on the bone surface. The proposed framework enables a more detailed investigation of the effect of disease and treatment on bones in preclinical studies and boosts the precision of such assessments.
Collapse
Affiliation(s)
- Stamatina Moraiti
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Vee San Cheong
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Future Health Technologies Programme, Singapore-ETH Centre, Create campus, Singapore, Singapore
| | - Enrico Dall’Ara
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Visakan Kadirkamanathan
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Pinaki Bhattacharya
- Department of Mechanical Engineering, University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
32
|
Tossetta G, Fantone S, Togni L, Santarelli A, Olivieri F, Marzioni D, Rippo MR. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants (Basel) 2024; 13:1270. [PMID: 39456522 PMCID: PMC11504014 DOI: 10.3390/antiox13101270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Periodontitis affects up to 40% of adults over 60 years old and is a consequence of gingivitis. Periodontitis is characterized by a chronic inflammation, periodontal damage, and alveolar bone resorption. The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling pathway plays a key role in periodontitis by modulating redox balance and inflammation of the periodontium. However, NRF2 expression is decreased in gingival tissues of patients with periodontitis while oxidative stress is significantly increased in this pathology. Oxidative stress and lipopolysaccharide (LPS) produced by gram-negative bacteria favor the production of inflammatory causing periodontal inflammation and favoring alveolar bone. In this review, we analyzed the current literature regarding the role of natural and synthetic compounds in modulating the NRF2/KEAP1 pathway in in vitro and in vivo models of periodontitis in order to evaluate new potential treatments of periodontitis that can improve the outcome of this disease.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Sonia Fantone
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (F.O.)
| | - Lucrezia Togni
- Department of Clinical Specialistic and Dental Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.T.); (A.S.)
| | - Andrea Santarelli
- Department of Clinical Specialistic and Dental Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.T.); (A.S.)
- Dentistry Clinic, National Institute of Health and Science of Aging, IRCCS INRCA, 60126 Ancona, Italy
| | - Fabiola Olivieri
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (F.O.)
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
- IRCCS INRCA, 60124 Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, 60126 Ancona, Italy;
| |
Collapse
|
33
|
Li B, Wu Y, Ying L, Zhu W, Yang J, Zhou L, Yi L, Jiang T, Jiang H, Song X, Xue W, Liang G, Huang S, Song Z. Synthesis and Antiosteoporotic Characterization of Diselenyl Maleimides: Discovery of a Potent Agent for the Treatment of Osteoporosis by Targeting RANKL. J Med Chem 2024; 67:17226-17242. [PMID: 39299698 DOI: 10.1021/acs.jmedchem.4c01105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
To discover new osteoclast-targeting antiosteoporosis agents, we identified forty-six diselenyl maleimides, which were efficiently prepared using a novel, simple, and metal-free method at room temperature in a short reaction time. Among them, 3k showed the most marked inhibition of osteoclast differentiation with an IC50 value of 0.36 ± 0.03 μM. Moreover, 3k significantly suppressed RANKL-induced osteoclast formation, bone resorption, and osteoclast-specific genes expression in vitro. Mechanistic studies revealed that 3k remarkably blocked the RANKL-induced mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways. In ovariectomized mice, intragastric administration of 3k significantly alleviated bone loss, exhibiting an effect similar to that of alendronate. Surface plasmon resonance assay and microscale thermophoresis assay results suggested that RANKL might be a potential molecular target for 3k. Collectively, the findings presented above provided a novel candidate for further development of bone antiresorptive drugs that target RANKL.
Collapse
Affiliation(s)
- Bin Li
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Yao Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Linkun Ying
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Zhu
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jingyi Yang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Lingling Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Lele Yi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Tianle Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Haofu Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Xiangrui Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Guang Liang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- School of Pharmacy, Hangzhou Medical College, Hangzhou 311399, Zhejiang, China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, No. 373, Xueyuan West Road, Lucheng District, Wenzhou 325027, PR China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| |
Collapse
|
34
|
Turker Yavas F, Sevil Kilimci F, Akkoc AN, Sahiner HS, Bardakci Yilmaz Ö. Melatonin's protective role against Bisphenol F and S-induced skeletal damage: A morphometric and histological study in rat. Ann Anat 2024; 256:152314. [PMID: 39053668 DOI: 10.1016/j.aanat.2024.152314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
This study aimed to evaluate the potential effects of Bisphenol F and S exposure on the skeletal structures of Sprague-Dawley rats. Given the increasing concern about the potential endocrine-disrupting effects of Bisphenol analogs on bone health, this research sought to elucidate their impact in conjunction with Melatonin. Using 80 male Sprague Dawley rats, bones were subjected to a 3-point bending test to assess mechanical properties, and histopathological evaluation was conducted after fixation and decalcification. Statistical analysis was performed using SPSS. The results of the mechanical tests revealed significant differences in deformation and elastic modulus values between groups treated with Bisphenol F+Melatonin and Bisphenol S+Melatonin compared to the control groups. However, the histological images showed no significant differences between the groups. In the discussion, it was noted that the injection of Bisphenol F and Melatonin together increased bone hardness, suggesting that Bisphenol F and Bisphenol S may mitigate the negative effects of melatonin on bone. We attributed the absence of histological differences to the male gender of the studied rats and previous exposure considerations. This study shows that Melatonin can reduce Bisphenol F and Bisphenol S' rapid adjustment effects and increase bone elasticity. The side effects of Bisphenol F and S, as well as the prophylactic effects of Melatonin, can be observed and improved by carefully adjusting the duration, dose, and gender selection.
Collapse
Affiliation(s)
- Firuze Turker Yavas
- Aydın Adnan Menderes University, Faculty of Veterinary Medicine, Department of Anatomy, Aydin 09016, Turkey.
| | - Figen Sevil Kilimci
- Aydın Adnan Menderes University, Faculty of Veterinary Medicine, Department of Anatomy, Aydin 09016, Turkey
| | - Ayse Nur Akkoc
- Aydın Adnan Menderes University, Faculty of Veterinary Medicine, Department of Pathology, Aydin 09016, Turkey
| | - Hande Sultan Sahiner
- Aydın Adnan Menderes University, Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Aydin 09016, Turkey
| | - Özge Bardakci Yilmaz
- Aydın Adnan Menderes University, Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Aydin 09016, Turkey
| |
Collapse
|
35
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
36
|
Wu Y, Sun B, Tang Y, Shen A, Lin Y, Zhao X, Li J, Monteiro MJ, Gu W. Bone targeted nano-drug and nano-delivery. Bone Res 2024; 12:51. [PMID: 39231955 PMCID: PMC11375042 DOI: 10.1038/s41413-024-00356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 09/06/2024] Open
Abstract
There are currently no targeted delivery systems to satisfactorily treat bone-related disorders. Many clinical drugs consisting of small organic molecules have a short circulation half-life and do not effectively reach the diseased tissue site. This coupled with repeatedly high dose usage that leads to severe side effects. With the advance in nanotechnology, drugs contained within a nano-delivery device or drugs aggregated into nanoparticles (nano-drugs) have shown promises in targeted drug delivery. The ability to design nanoparticles to target bone has attracted many researchers to develop new systems for treating bone related diseases and even repurposing current drug therapies. In this review, we shall summarise the latest progress in this area and present a perspective for future development in the field. We will focus on calcium-based nanoparticle systems that modulate calcium metabolism and consequently, the bone microenvironment to inhibit disease progression (including cancer). We shall also review the bone affinity drug family, bisphosphonates, as both a nano-drug and nano-delivery system for bone targeted therapy. The ability to target and release the drug in a controlled manner at the disease site represents a promising safe therapy to treat bone diseases in the future.
Collapse
Affiliation(s)
- Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Ying Tang
- Science and Technology Innovation Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aining Shen
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yanlin Lin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Xiaohui Zhao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Centre for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
37
|
Sun Q, Xu L, Hu Z, Liu J, Yu T, Li M, Zhang S, Shi F. Melatonin Regulates Osteoblast Differentiation through the m6A Reader hnRNPA2B1 under Simulated Microgravity. Curr Issues Mol Biol 2024; 46:9624-9638. [PMID: 39329924 PMCID: PMC11430354 DOI: 10.3390/cimb46090572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Recent studies have confirmed that melatonin and N6-methyladenosine (m6A) modification can influence bone cell differentiation and bone formation. Melatonin can also regulate a variety of biological processes through m6A modification. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1) serves as a reader of m6A modification. In this study, we used the hindlimb unloading model as an animal model of bone loss induced by simulated microgravity and used 2D clinorotation to simulate a microgravity environment for cells on the ground. We found that hnRNPA2B1 was downregulated both in vitro and in vivo during simulated microgravity. Further investigations showed that hnRNPA2B1 could promote osteoblast differentiation and that overexpression of hnRNPA2B1 attenuated the suppression of osteoblast differentiation induced by simulated microgravity. We also discovered that melatonin could promote the expression of hnRNPA2B1 under simulated microgravity. Moreover, we found that promotion of osteoblast differentiation by melatonin was partially dependent on hnRNPA2B1. Therefore, this research revealed, for the first time, the role of the melatonin/hnRNPA2B1 axis in osteoblast differentiation under simulated microgravity. Targeting this axis may be a potential protective strategy against microgravity-induced bone loss and osteoporosis.
Collapse
Affiliation(s)
- Quan Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Liqun Xu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Jingchun Liu
- No. 5 Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi’an 710032, China; (J.L.); (T.Y.)
| | - Tingfei Yu
- No. 5 Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi’an 710032, China; (J.L.); (T.Y.)
| | - Meng Li
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi’an 710032, China; (Q.S.); (L.X.); (Z.H.); (M.L.)
| |
Collapse
|
38
|
Prins CM, Ceylan M, Hogervorst JMA, Jansen IDC, Schimmel IM, Schoenmaker T, de Vries TJ. Osteogenic differentiation of periodontal ligament fibroblasts inhibits osteoclast formation. Eur J Cell Biol 2024; 103:151440. [PMID: 38954934 DOI: 10.1016/j.ejcb.2024.151440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/02/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
One of the deficits of knowledge on bone remodelling, is to what extent cells that are driven towards osteogenic differentiation can contribute to osteoclast formation. The periodontal ligament fibroblast (PdLFs) is an ideal model to study this, since they play a role in osteogenesis, and can also orchestrate osteoclastogenesis.when co-cultured with a source of osteoclast-precursor such as peripheral blood mononuclear cells (PBMCs). Here, the osteogenic differentiation of PdLFs and the effects of this process on the formation of osteoclasts were investigated. PdLFs were obtained from extracted teeth and exposed to osteogenic medium for 0, 7, 14, or 21 out of 21 days. After this 21-day culturing period, the cells were co-cultured with peripheral blood mononuclear cells (PBMCs) for an additional 21 days to study osteoclast formation. Alkaline phosphatase (ALP) activity, calcium concentration, and gene expression of osteogenic markers were assessed at day 21 to evaluate the different stages of osteogenic differentiation. Alizarin red staining and scanning electron microscopy were used to visualise mineralisation. Tartrate-resistant acid phosphatase (TRAcP) activity, TRAcP staining, multinuclearity, the expression of osteoclastogenesis-related genes, and TNF-α and IL-1β protein levels were assessed to evaluate osteoclastogenesis. The osteogenesis assays revealed that PdLFs became more differentiated as they were exposed to osteogenic medium for a longer period of time. Mineralisation by these osteogenic cells increased with the progression of differentiation. Culturing PdLFs in osteogenic medium before co-culturing them with PMBCs led to a significant decrease in osteoclast formation. qPCR revealed significantly lower DCSTAMP expression in cultures that had been supplemented with osteogenic medium. Protein levels of osteoclastogenesis stimulator TNF-α were also lower in these cultures. The present study shows that the osteogenic differentiation of PdLFs reduces the osteoclastogenic potential of these cells. Immature cells of the osteoblastic lineage may facilitate osteoclastogenesis, whereas mature mineralising cells may suppress the formation of osteoclasts. Therefore, mature and immature osteogenic cells may have different roles in maintaining bone homeostasis.
Collapse
Affiliation(s)
- Caya M Prins
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands; Amsterdam University College (University of Amsterdam and Vrije Universiteit), Amsterdam, the Netherlands
| | - Merve Ceylan
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Ineke D C Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Irene M Schimmel
- Department of Medical Biology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands.
| |
Collapse
|
39
|
Feng X, Wei G, Su Y, Xian Y, Liu Z, Gao Y, Liang J, Lian H, Xu J, Zhao J, Liu Q, Song F. Active fraction of Polyrhachis vicina (Rogers) inhibits osteoclastogenesis by targeting Trim38 mediated proteasomal degradation of TRAF6. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155890. [PMID: 39033726 DOI: 10.1016/j.phymed.2024.155890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/25/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Reactive Oxygen Species (ROS) is a key factor in the pathogenesis of osteoporosis (OP) primarily characterized by excessive osteoclast activity. Active fraction of Polyrhachis vicina Rogers (AFPR) exerts antioxidant effects and possesses extensive promising therapeutic effects in various conditions, however, its function in osteoclastogenesis and OP is unknown. PURPOSE The aim of this study is to elucidate the cellular and molecular mechanisms of AFPR in OP. STUDY DESIGN AND METHODS CCK8 assay was used to evaluate the cell viability under AFPR treatment. TRAcP staining, podosome belts staining and bone resorption were used to test the effect of AFPR on osteoclastogenesis. Immunofluorescence staining was used to observe the effect of AFPR on ROS production. si-RNA transfection, coimmunoprecipitation and Western-blot were used to clarify the underlying mechanisms. Further, an ovariectomy (OVX) -induced OP mice model was used to identify the effect of AFPR on bone loss using Micro-CT scanning and histological examination. RESULTS In the present study, AFPR inhibited osteoclast differentiation and bone resorption induced by nuclear factor-κB receptor activator (NF-κB) ligand (RANKL) in dose-/ time-dependent with no cytotoxicity. Meanwhile, AFPR decreased RANKL-mediated ROS levels and enhanced ROS scavenging enzymes. Mechanistically, AFPR promoted proteasomal degradation of TRAF6 by significantly upregulating its K48-linked ubiquitination, subsequently inhibiting NFATc1 activity. We further observed that tripartite motif protein 38 (TRIM38) could mediate the ubiquitination of TRAF6 in response to RANKL. Moreover, TRIM38 could negatively regulate the RANKL pathway by binding to TRAF6 and promoting K48-linked polyubiquitination. In addition, TRIM38 deficiency rescued the inhibition of AFPR on ROS and NFATc1 activity and osteoclastogenesis. In line with these results, AFPR reduced OP caused by OVX through ameliorating osteoclastogenesis. CONCLUSION AFPR alleviates ovariectomized-induced bone loss via suppressing ROS and NFATc1 by targeting Trim38 mediated proteasomal degradation of TRAF6. The research offers innovative perspectives on AFPR's suppressive impact in vivo OVX mouse model and in vitro, and clarifies the fundamental mechanism.
Collapse
Affiliation(s)
- Xiaoliang Feng
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Guining Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, PR China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yansi Xian
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zhijuan Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yijie Gao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jiamin Liang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Haoyu Lian
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, Australia; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China.
| | - Fangming Song
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China.
| |
Collapse
|
40
|
Hu W, Chen S, Zou X, Chen Y, Luo J, Zhong P, Ma D. Oral microbiome, periodontal disease and systemic bone-related diseases in the era of homeostatic medicine. J Adv Res 2024:S2090-1232(24)00362-X. [PMID: 39159722 DOI: 10.1016/j.jare.2024.08.019] [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: 03/18/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Homeostasis is a state of self-regulation and dynamic equilibrium, maintaining the good physiological functions of each system in living organisms. In the oral cavity, the interaction between the host and the oral microbiome forms oral microbial homeostasis. Physiological bone remodeling and renewal can occur under the maintenance of oral microbial homeostasis. The imbalance of bone homeostasis is a key mechanism leading to the occurrence of systemic bone-related diseases. Considering the importance of oral microbial homeostasis in the maintenance of bone homeostasis, it still lacks a complete understanding of the relationship between oral microbiome, periodontal disease and systemic bone-related diseases. AIM OF REVIEW This review focuses on the homeostatic changes, pathogenic routes and potential mechanisms in the oral microbiome in periodontal disease and systemic bone-related diseases such as rheumatoid arthritis, osteoarthritis, osteoporosis and osteomyelitis. Additionally, this review discusses oral microbiome-based diagnostic approaches and explores probiotics, mesenchymal stem cells, and oral microbiome transplantation as promising treatment strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the association between oral microbial homeostasis imbalance and systemic bone-related diseases, and highlights the possibility of remodeling oral microbial homeostasis for the prevention and treatment of systemic bone-related diseases.
Collapse
Affiliation(s)
- Weiqi Hu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Shuoling Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Xianghui Zou
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Yan Chen
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Jiayu Luo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Peiliang Zhong
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Dandan Ma
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China.
| |
Collapse
|
41
|
An JY, Ma XN, Wen HL, Hu HD. Identification of key genes and long non‑coding RNA expression profiles in osteoporosis with rheumatoid arthritis based on bioinformatics analysis. BMC Musculoskelet Disord 2024; 25:634. [PMID: 39118036 PMCID: PMC11312199 DOI: 10.1186/s12891-024-07738-x] [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: 07/21/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Although rheumatoid arthritis (RA) is a chronic systemic tissue disease often accompanied by osteoporosis (OP), the molecular mechanisms underlying this association remain unclear. This study aimed to elucidate the pathogenesis of RA and OP by identifying differentially expressed mRNAs (DEmRNAs) and long non-coding RNAs (lncRNAs) using a bioinformatics approach. METHODS Expression profiles of individuals diagnosed with OP and RA were retrieved from the Gene Expression Omnibus database. Differential expression analysis was conducted. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) pathway enrichment analyses were performed to gain insights into the functional categories and molecular/biochemical pathways associated with DEmRNAs. We identified the intersection of common DEmRNAs and lncRNAs and constructed a protein-protein interaction (PPI) network. Correlation analysis between the common DEmRNAs and lncRNAs facilitated the construction of a coding-non-coding network. Lastly, serum peripheral blood mononuclear cells (PBMCs) from patients with RA and OP, as well as healthy controls, were obtained for TRAP staining and qRT-PCR to validate the findings obtained from the online dataset assessments. RESULTS A total of 28 DEmRNAs and 2 DElncRNAs were identified in individuals with both RA and OP. Chromosomal distribution analysis of the consensus DEmRNAs revealed that chromosome 1 had the highest number of differential expression genes. GO and KEGG analyses indicated that these DEmRNAs were primarily associated with " platelets (PLTs) degranulation", "platelet alpha granules", "platelet activation", "tight junctions" and "leukocyte transendothelial migration", with many genes functionally related to PLTs. In the PPI network, MT-ATP6 and PTGS1 emerged as potential hub genes, with MT-ATP6 originating from mitochondrial DNA. Co-expression analysis identified two key lncRNA-mRNA pairs: RP11 - 815J21.2 with MT - ATP6 and RP11 - 815J21.2 with PTGS1. Experimental validation confirmed significant differential expression of RP11-815J21.2, MT-ATP6 and PTGS1 between the healthy controls and the RA + OP groups. Notably, knockdown of RP11-815J21.2 attenuated TNF + IL-6-induced osteoclastogenesis. CONCLUSIONS This study successfully identified shared dysregulated genes and potential therapeutic targets in individuals with RA and OP, highlighting their molecular similarities. These findings provide new insights into the pathogenesis of RA and OP and suggest potential avenues for further research and targeted therapies.
Collapse
Affiliation(s)
- Jin-Yu An
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China.
| | - Xing-Na Ma
- Department of Pediatric, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| | - Hui-Long Wen
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| | - Hui-Dong Hu
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| |
Collapse
|
42
|
Chen Y, Xiao H, Liu Z, Teng F, Yang A, Geng B, Sheng X, Xia Y. Sirt1: An Increasingly Interesting Molecule with a Potential Role in Bone Metabolism and Osteoporosis. Biomolecules 2024; 14:970. [PMID: 39199358 PMCID: PMC11352324 DOI: 10.3390/biom14080970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Osteoporosis (OP) is a common metabolic bone disease characterized by low bone mass, decreased bone mineral density, and degradation of bone tissue microarchitecture. However, our understanding of the mechanisms of bone remodeling and factors affecting bone mass remains incomplete. Sirtuin1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent deacetylase that regulates a variety of cellular metabolisms, including inflammation, tumorigenesis, and bone metabolism. Recent studies have emphasized the important role of SIRT1 in bone homeostasis. This article reviews the role of SIRT1 in bone metabolism and OP and also discusses therapeutic strategies and future research directions for targeting SIRT1.
Collapse
Affiliation(s)
- Yi Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Hefang Xiao
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Zirui Liu
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Fei Teng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Ao Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Xiaoyun Sheng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
43
|
Li X, Tian H, Zhang J, Dou B. Zoledronic Acid and Enriched Autologous Bone Marrow Stem Cell Implantation for Femoral Head Osteonecrosis. Indian J Orthop 2024; 58:1064-1069. [PMID: 39087050 PMCID: PMC11286889 DOI: 10.1007/s43465-024-01188-2] [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] [Received: 09/16/2023] [Accepted: 05/16/2024] [Indexed: 08/02/2024]
Abstract
Purpose This study evaluated the clinical results of zoledronic acid in the treatment of early osteonecrosis of the femoral head (ONFH). Materials and Methods Study retrospectively analyzed 60 patients with zoledronic acid with bone marrow stem cell (BMSC) implantation (The study group) and 64 patients with BMSC implantation (The control group). The primary evaluation index included VAS, HHS, collapsed rate, and total hip replacement arthroplasty (THA) conversion rate. Results The study group had a lower VAS (1.12 ± 0.22 vs 1.44 ± 0.32) and higher HHS (75.07 ± 3.66 vs 68.78 ± 2.24) compared to the control group in 6 months after surgery (P < 0.05). In the study group, 12 hips (20%) collapsed, and 7 of 60 hips (11.67%) required THA surgery at the last follow-up. However, 25 hips (38.8%) collapsed in the control group, and 19 hips (29.69%) required THA surgery. The study group had a lower collapsed rate (P = 0.029) and THA conversion rate (P = 0.016) in survival analysis. Conclusion Zoledronic acid and BMSC implantation in the treatment of early ONFH is safe and effective, reduces pain shortens recovery time, and reduces collapsed rate and THA conversion rate in ONFH patients.
Collapse
Affiliation(s)
- Xuedong Li
- Zhengzhou Orthopaedic Hospital, Longhai Middle Road 58, Zhengzhou, 450000 Henan China
| | | | - Jingyi Zhang
- Zhengzhou Orthopaedic Hospital, Longhai Middle Road 58, Zhengzhou, 450000 Henan China
| | - Bangxian Dou
- Zhengzhou Orthopaedic Hospital, Longhai Middle Road 58, Zhengzhou, 450000 Henan China
| |
Collapse
|
44
|
Cao M, Sheng R, Sun Y, Cao Y, Wang H, Zhang M, Pu Y, Gao Y, Zhang Y, Lu P, Teng G, Wang Q, Rui Y. Delivering Microrobots in the Musculoskeletal System. NANO-MICRO LETTERS 2024; 16:251. [PMID: 39037551 PMCID: PMC11263536 DOI: 10.1007/s40820-024-01464-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/16/2024] [Indexed: 07/23/2024]
Abstract
Disorders of the musculoskeletal system are the major contributors to the global burden of disease and current treatments show limited efficacy. Patients often suffer chronic pain and might eventually have to undergo end-stage surgery. Therefore, future treatments should focus on early detection and intervention of regional lesions. Microrobots have been gradually used in organisms due to their advantages of intelligent, precise and minimally invasive targeted delivery. Through the combination of control and imaging systems, microrobots with good biosafety can be delivered to the desired area for treatment. In the musculoskeletal system, microrobots are mainly utilized to transport stem cells/drugs or to remove hazardous substances from the body. Compared to traditional biomaterial and tissue engineering strategies, active motion improves the efficiency and penetration of local targeting of cells/drugs. This review discusses the frontier applications of microrobotic systems in different tissues of the musculoskeletal system. We summarize the challenges and barriers that hinder clinical translation by evaluating the characteristics of different microrobots and finally point out the future direction of microrobots in the musculoskeletal system.
Collapse
Affiliation(s)
- Mumin Cao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Renwang Sheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yimin Sun
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ying Cao
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yunmeng Pu
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yucheng Gao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Gaojun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Qianqian Wang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China.
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
45
|
Meng X, Wang WD, Li SR, Sun ZJ, Zhang L. Harnessing cerium-based biomaterials for the treatment of bone diseases. Acta Biomater 2024; 183:30-49. [PMID: 38849022 DOI: 10.1016/j.actbio.2024.05.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Bone, an actively metabolic organ, undergoes constant remodeling throughout life. Disturbances in the bone microenvironment can be responsible for pathologically bone diseases such as periodontitis, osteoarthritis, rheumatoid arthritis and osteoporosis. Conventional bone tissue biomaterials are not adequately adapted to complex bone microenvironment. Therefore, there is an urgent clinical need to find an effective strategy to improve the status quo. In recent years, nanotechnology has caused a revolution in biomedicine. Cerium(III, IV) oxide, as an important member of metal oxide nanomaterials, has dual redox properties through reversible binding with oxygen atoms, which continuously cycle between Ce(III) and Ce(IV). Due to its special physicochemical properties, cerium(III, IV) oxide has received widespread attention as a versatile nanomaterial, especially in bone diseases. This review describes the characteristics of bone microenvironment. The enzyme-like properties and biosafety of cerium(III, IV) oxide are also emphasized. Meanwhile, we summarizes controllable synthesis of cerium(III, IV) oxide with different nanostructural morphologies. Following resolution of synthetic principles of cerium(III, IV) oxide, a variety of tailored cerium-based biomaterials have been widely developed, including bioactive glasses, scaffolds, nanomembranes, coatings, and nanocomposites. Furthermore, we highlight the latest advances in cerium-based biomaterials for inflammatory and metabolic bone diseases and bone-related tumors. Tailored cerium-based biomaterials have already demonstrated their value in disease prevention, diagnosis (imaging and biosensors) and treatment. Therefore, it is important to assist in bone disease management by clarifying tailored properties of cerium(III, IV) oxide in order to promote the use of cerium-based biomaterials in the future clinical setting. STATEMENT OF SIGNIFICANCE: In this review, we focused on the promising of cerium-based biomaterials for bone diseases. We reviewed the key role of bone microenvironment in bone diseases and the main biological activities of cerium(III, IV) oxide. By setting different synthesis conditions, cerium(III, IV) oxide nanostructures with different morphologies can be controlled. Meanwhile, tailored cerium-based biomaterials can serve as a versatile toolbox (e.g., bioactive glasses, scaffolds, nanofibrous membranes, coatings, and nanocomposites). Then, the latest research advances based on cerium-based biomaterials for the treatment of bone diseases were also highlighted. Most importantly, we analyzed the perspectives and challenges of cerium-based biomaterials. In future perspectives, this insight has given rise to a cascade of cerium-based biomaterial strategies, including disease prevention, diagnosis (imaging and biosensors) and treatment.
Collapse
Affiliation(s)
- Xiang Meng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Su-Ran Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China.
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China; Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan, 430079, PR China.
| |
Collapse
|
46
|
Galchenko A, Rizzo G, Sidorova E, Skliar E, Baroni L, Visaggi P, Guidi G, de Bortoli N. Bone mineral density parameters and related nutritional factors in vegans, lacto-ovo-vegetarians, and omnivores: a cross-sectional study. Front Nutr 2024; 11:1390773. [PMID: 38919395 PMCID: PMC11196821 DOI: 10.3389/fnut.2024.1390773] [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: 02/23/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction The growing prevalence of vegetarianism determines the need for comprehensive study of the impact of these diets on health and particularly on bone metabolism. We hypothesized that significant dietary differences between vegans, lacto-ovo-vegetarians, and omnivores also cause significant differences in their nutrient status, which may affect bone health. Methods The study assessed dual-energy X-ray absorptiometry parameters in lumbar spine and femoral neck, average nutrient intake, serum nutrient concentrations, serum PTH levels, and urinary pH among 46 vegans, 38 lacto-ovo-vegetarians, and 44 omnivores. Results There were no differences in bone mineral density (BMD) between the groups. However, the parathyroid hormone (PTH) levels were still higher in vegans compared to omnivores, despite the same prevalence of hyperparathyroidism in all groups. These findings may probably be explained by the fact that each group had its own "strengths and weaknesses." Thus, vegans and, to a lesser extent, lacto-ovo-vegetarians consumed much more potassium, magnesium, copper, manganese, and vitamins B6, B9, and C. At the same time, the diet of omnivores contained more protein and vitamins D and B12. All the subjects consumed less vitamin D than recommended. More than half of vegans and omnivores had insufficiency or even deficiency of vitamin D in the blood. Low serum concentrations of manganese with its quite adequate intake are also noteworthy: its deficiency was observed in 57% of vegans, 79% of lacto-ovo-vegetarians, and 63% of omnivores. Discussion Currently, it is no longer possible to conclude that lacto-ovo-vegetarians have lower BMD than omnivores, as our research supported. Vegans in our study also did not demonstrate lower BMD values, only higher PTH blood concentrations, compared to omnivores, however, a large number of studies, including recent, show the opposite view. In this regard, further large-scale research is required. Vegans and lacto-ovo-vegetarians now have a variety of foods fortified with vitamins D and B12, as well as calcium. There is also a great diversity of ethically sourced dietary supplements. The found low concentrations of manganese require further investigation.
Collapse
Affiliation(s)
- Alexey Galchenko
- Scientific Society for Vegetarian Nutrition, Venice, Italy
- Earth Philosophical Society “Melodia Vitae”, International, Toronto, CA, Canada
| | | | | | - Elena Skliar
- Earth Philosophical Society “Melodia Vitae”, International, Toronto, CA, Canada
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Luciana Baroni
- Scientific Society for Vegetarian Nutrition, Venice, Italy
| | - Pierfrancesco Visaggi
- Division of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giada Guidi
- Division of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Nicola de Bortoli
- Division of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- NUTRAFOOD, Interdepartmental Center for Nutraceutical Research and Nutrition for Health, University of Pisa, Pisa, Italy
| |
Collapse
|
47
|
Li W, Niu Y, Qiu Z, Zhou S, Zhong W, Xiong Z, Zhao D, Yang Y, Zhao H, Yu X. New evidence on the controversy over the correlation between vertebral osteoporosis and intervertebral disc degeneration: a systematic review of relevant animal studies. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:2354-2379. [PMID: 38642137 DOI: 10.1007/s00586-024-08256-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVE The effect of vertebral osteoporosis on disc degeneration remains controversial. The aim of this study was to conduct a systematic review and meta-analysis of relevant animal studies to shed more light on the effects and mechanisms of vertebral osteoporosis on disc degeneration and to promote the resolution of the controversy. METHODS The PubMed, Cochrane Library, and Embase databases were searched for studies that met the inclusion criteria. Basic information and data were extracted from the included studies and data were analyzed using STATA 15.1 software. This study was registered on INPLASY with the registration number INPLASY202370099 and https://doi.org/10.37766/inplasy2023.7.0099 . RESULTS A total of 13 studies were included in our study. Both animals, rats and mice, were covered. Meta-analysis results showed in disc height index (DHI) (P < 0.001), histological score (P < 0.001), number of osteoblasts in the endplate (P = 0.043), number of osteoclasts in the endplate (P < 0.001), type I collagen (P < 0.001), type II collagen (P < 0.001), aggrecan (P < 0.001), recombinant a disintegrin and metalloproteinase with thrombospondin-4 (ADAMTS-4) (P < 0.001), matrix metalloproteinase-1 (MMP-1) (P < 0.001), MMP-3 (P < 0.001), MMP-13 (P < 0.001), the difference between the osteoporosis group and the control group was statistically significant. In terms of disc volume, the difference between the osteoporosis group and the control group was not statistically significant (P = 0.459). CONCLUSION Our study shows that vertebral osteoporosis may exacerbate disc degeneration. Abnormal bone remodeling caused by vertebral osteoporosis disrupts the structural integrity of the endplate, leading to impaired nutrient supply to the disc, increased expression of catabolic factors, and decreased levels of type II collagen and aggrecan may be one of the potential mechanisms.
Collapse
Affiliation(s)
- Wenhao Li
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yiqun Niu
- China Medical Technology Press Co., Ltd, Beijing, 100089, China
| | - Ziye Qiu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Shibo Zhou
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Wenqing Zhong
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Zhencheng Xiong
- West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Dingyan Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yongdong Yang
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - He Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Xing Yu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
48
|
Li T, Du Y, Yao H, Zhao B, Wang Z, Chen R, Ji Y, Du M. Isobavachin attenuates osteoclastogenesis and periodontitis-induced bone loss by inhibiting cellular iron accumulation and mitochondrial biogenesis. Biochem Pharmacol 2024; 224:116202. [PMID: 38615917 DOI: 10.1016/j.bcp.2024.116202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/24/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
As bone-resorbing cells rich in mitochondria, osteoclasts require high iron uptake to promote mitochondrial biogenesis and maintain a high-energy metabolic state for active bone resorption. Given that abnormal osteoclast formation and activation leads to imbalanced bone remodeling and osteolytic bone loss, osteoclasts may be crucial targets for treating osteolytic diseases such as periodontitis. Isobavachin (IBA), a natural flavonoid compound, has been confirmed to be an inhibitor of receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation from bone marrow-derived macrophages (BMMs). However, its effects on periodontitis-induced bone loss and the potential mechanism of its anti-osteoclastogenesis effect remain unclear. Our study demonstrated that IBA suppressed RANKL-induced osteoclastogenesis in BMMs and RAW264.7 cells and inhibited osteoclast-mediated bone resorption in vitro. Transcriptomic analysis indicated that iron homeostasis and reactive oxygen species (ROS) metabolic process were enriched among the differentially expressed genes following IBA treatment. IBA exerted its anti-osteoclastogenesis effect by inhibiting iron accumulation in osteoclasts. Mechanistically, IBA attenuated iron accumulation in RANKL-induced osteoclasts by inhibiting the mitogen-activated protein kinase (MAPK) pathway to upregulate ferroportin1 (Fpn1) expression and promote Fpn1-mediated intracellular iron efflux. We also found that IBA inhibited mitochondrial biogenesis and function, and reduced RANKL-induced ROS generation in osteoclasts. Furthermore, IBA attenuated periodontitis-induced bone loss by reducing osteoclastogenesis in vivo. Overall, these results suggest that IBA may serve as a promising therapeutic strategy for bone diseases characterized by osteoclastic bone resorption.
Collapse
Affiliation(s)
- Ting Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yangge Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hantao Yao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Boxuan Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zijun Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rourong Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaoting Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Minquan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
49
|
Cheng X, Tian W, Yang J, Wang J, Zhang Y. Engineering approaches to manipulate osteoclast behavior for bone regeneration. Mater Today Bio 2024; 26:101043. [PMID: 38600918 PMCID: PMC11004223 DOI: 10.1016/j.mtbio.2024.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Extensive research has delved into the multifaceted roles of osteoclasts beyond their traditional function in bone resorption in recent years, uncovering their significant influence on bone formation. This shift in understanding has spurred investigations into engineering strategies aimed at leveraging osteoclasts to not only inhibit bone resorption but also facilitate bone regeneration. This review seeks to comprehensively examine the mechanisms by which osteoclasts impact bone metabolism. Additionally, it explores various engineering methodologies, including the modification of bioactive material properties, localized drug delivery, and the introduction of exogenous cells, assessing their potential and mechanisms in aiding bone repair by targeting osteoclasts. Finally, the review proposes current limitations and future routes for manipulating osteoclasts through biological and material cues to facilitate bone repair.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, 1098 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
| | - Wenzhi Tian
- Jilin University, Jilin Province Key Lab Tooth Dev & Bone Remodeling, School and Hospital of Stomatology, Department of Oral Pathology, Changchun 130041, Jilin Province, China
| | - Jianhua Yang
- Longgang District People's Hospital of Shenzhen & the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, Guangdong province, China
| | - Jiamian Wang
- National Innovation Center for Advanced Medical Devices, Shenzhen 518000, Guangdong Province, China
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, 1088 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
- School of Biomedical Engineering, Shenzhen University Medical School, 1088 Xueyuan Road, Shenzhen 518055, Guangdong Province, China
| |
Collapse
|
50
|
Kim M, Park JH, Go M, Lee N, Seo J, Lee H, Kim D, Ha H, Kim T, Jeong MS, Kim S, Kim T, Kim HS, Kang D, Shim H, Lee SY. RUFY4 deletion prevents pathological bone loss by blocking endo-lysosomal trafficking of osteoclasts. Bone Res 2024; 12:29. [PMID: 38744829 PMCID: PMC11094054 DOI: 10.1038/s41413-024-00326-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 05/16/2024] Open
Abstract
Mature osteoclasts degrade bone matrix by exocytosis of active proteases from secretory lysosomes through a ruffled border. However, the molecular mechanisms underlying lysosomal trafficking and secretion in osteoclasts remain largely unknown. Here, we show with GeneChip analysis that RUN and FYVE domain-containing protein 4 (RUFY4) is strongly upregulated during osteoclastogenesis. Mice lacking Rufy4 exhibited a high trabecular bone mass phenotype with abnormalities in osteoclast function in vivo. Furthermore, deleting Rufy4 did not affect osteoclast differentiation, but inhibited bone-resorbing activity due to disruption in the acidic maturation of secondary lysosomes, their trafficking to the membrane, and their secretion of cathepsin K into the extracellular space. Mechanistically, RUFY4 promotes late endosome-lysosome fusion by acting as an adaptor protein between Rab7 on late endosomes and LAMP2 on primary lysosomes. Consequently, Rufy4-deficient mice were highly protected from lipopolysaccharide- and ovariectomy-induced bone loss. Thus, RUFY4 plays as a new regulator in osteoclast activity by mediating endo-lysosomal trafficking and have a potential to be specific target for therapies against bone-loss diseases such as osteoporosis.
Collapse
Affiliation(s)
- Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Jin Hee Park
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea
| | - Miyeon Go
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Nawon Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Jeongin Seo
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Hyunil Ha
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, South Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, South Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, South Korea
| | - Suree Kim
- Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Taesoo Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Dongmin Kang
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyunbo Shim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea.
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, 03760, South Korea.
| |
Collapse
|