1
|
Chen L, Zhu J, Ge N, Liu Y, Yan Z, Liu G, Li Y, Wang Y, Wu G, Qiu T, Dai H, Han J, Guo C. A biodegradable magnesium alloy promotes subperiosteal osteogenesis via interleukin-10-dependent macrophage immunomodulation. Biomaterials 2025; 318:122992. [PMID: 39862617 DOI: 10.1016/j.biomaterials.2024.122992] [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: 05/16/2024] [Revised: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 01/27/2025]
Abstract
In situ bone regeneration and vertical bone augmentation have been huge problems in clinical practice, always imposing a significant economic burden and causing patient suffering. Herein, MgZnYNd magnesium alloy rod implantation in mouse femur resulted in substantial subperiosteal new bone formation, with osteoimmunomodulation playing a pivotal role. Abundant macrophages were attracted to the subperiosteal new bone region and proved to be the most important regulation cells for bone regeneration. Periosteum stripping, macrophage depletion, and interleukin-10 (IL-10) blockade effectively diminished the MgZnYNd alloy-induced subperiosteal osteogenesis. Mechanistically, the degradation products of MgZnYNd alloy promoted M2 macrophage polarization and the secretion of anti-inflammatory cytokine IL-10, which enhanced periosteum-derived stem cells (PDSCs) osteogenesis through the JAK1-STAT3 pathway. An anti-IL-10 neutralizing antibody or STAT3 inhibitor significantly inhibited M2 macrophage-mediated osteogenic differentiation of PDSCs. Transcriptomics and proteomics revealed that periostin is the core regulator of PDSCs osteogenic differentiation. Furthermore, a novel clinical translation application of Mg-induced subperiosteal osteogenesis was developed, demonstrating its ability to preserve the height and width of the alveolar crest in rats and rabbits following tooth extraction. Collectively, these findings unveil a previously undefined role for Mg alloy-induced subperiosteal osteogenesis via macrophage-mediated osteoimmunomodulation, suggesting the therapeutic potential of magnesium alloy in bone regeneration and bone augmentation.
Collapse
Affiliation(s)
- Liangwei Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Jianhua Zhu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Na Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yan Liu
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China; Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ziyu Yan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guanqi Liu
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yuqi Li
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yifei Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guanxi Wu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Tiancheng Qiu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
| | - Jianmin Han
- Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China.
| | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China; Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China.
| |
Collapse
|
2
|
Su S, Bai J, Wang R, Gao S, Zhou R, Zhou F. A novel strategy for bone defect repair: Stromal cell-derived factor 1α sustained-release acellular fish scale scaffolds combined with injection of bone marrow mesenchymal stem cells promote bone regeneration. Mater Today Bio 2025; 32:101759. [PMID: 40270891 PMCID: PMC12017916 DOI: 10.1016/j.mtbio.2025.101759] [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: 02/25/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Patients with bone defects often have weak cell vitality and differentiation ability of endogenous bone marrow mesenchymal stem cells (BMSCs), which makes bone regeneration face challenges. At present, the bone tissue engineering strategies are mainly to build grafts by loading cells on scaffolds in vitro. These strategies face many difficulties that limit their clinical application. To this end, we developed a new strategy for bone defect repair, namely chemotactic cell-free scaffolds combined with BMSCs injection. We first prepared a polydopamine-functionalized acellular fish scale scaffold that can continuously release stromal cell-derived factor 1α (SDF-1α) (termed as SDF-1α/PAFS) in vivo for at least 10 days. The study results showed that the scaffold not only has excellent mechanical properties and good biocompatibility but also has reactive oxygen scavenging activity, immunomodulation, angiogenesis, and osteogenesis. More importantly, SDF-1α/PAFS can recruit postoperatively injected BMSCs into bone defects for bone repair. We constructed the mouse cranial bone defect model, and in vivo experimental results confirmed that the strategy of combining SDF-1α/PAFS with BMSCs injection can effectively promote bone defect repair. Overall, this study provides a promising strategy for bone defect repair, with better clinical convenience and operability.
Collapse
Affiliation(s)
- Shilong Su
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Shan Gao
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| | - Rubing Zhou
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No.95 Yong'an Road, Xicheng, 100050, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian, 100191, Beijing, China
| |
Collapse
|
3
|
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
|
4
|
Zhang X, Sun X, Luo Y, Wang X, Mao C, Xu Z, Song Y, Yan J. Endogenous sulfur dioxide deficiency impairs bone regeneration through abolishing S-sulfenylating p38 at cysteine 211. Int Immunopharmacol 2025; 158:114814. [PMID: 40347882 DOI: 10.1016/j.intimp.2025.114814] [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: 09/22/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Bone defects result in substantial medical expenses and a diminished quality of life for patients. Macrophage polarization is crucial in the bone regeneration process mediated by bone marrow-derived mesenchymal stem cells (BMSCs). macrophage-derived sulfur dioxide (SO2), the fourth endogenous gas signaling molecule, following hydrogen sulfide (H2S), has been shown to regulate macrophage chemotaxis and inflammatory responses. Nevertheless, the specific regulatory effects and mechanisms of macrophage-derived SO2 on bone regeneration are not yet fully understood. This study reveals for the first time that the absence of macrophage-derived SO2 promotes M1 macrophage polarization, whereas the administration of exogenous SO2 donors inhibits M1 polarization. The deficiency of macrophage-derived SO2 results in impaired osteogenic differentiation of BMSCs, whereas the administration of SO2 donors enhances this differentiation process. Further investigations have elucidated that p38α MAPK (p38) is crucial in mediating SO2's effects on M1 macrophage polarization and BMSCs osteogenic differentiation. Mechanistically, SO2 induces S-sulfenylation of p38 in macrophages, an effect that can be reversed by the thiol reductant dithiothreitol. Additionally, the C211S mutation in p38 abrogates the SO2-induced S-sulfenylation of p38, thereby preventing the inhibition of p38 activation and subsequently disrupting the regulation of M1 macrophage polarization and BMSCs osteogenic differentiation. In a model of mouse calvarial bone defects, we consistently observed that inhibiting SO2 production using the SO2-generating enzyme inhibitor HDX impaired bone regeneration capacity in mice, whereas the administration of an SO2 donor enhanced this capacity. In summary, macrophage-derived SO2 S-sulfenylates p38 at cysteine 211, thereby suppressing p38 activation, which inhibits M1 polarization and subsequently maintains the osteogenic differentiation of BMSCs. This study is the first to elucidate the role and mechanism of SO2 in sustaining osteogenesis, offering a novel strategy for addressing bone defect-related disorders.
Collapse
Affiliation(s)
- Xuanming Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xutao Sun
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yumeng Luo
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaoyan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Caiyun Mao
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zihang Xu
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yunjia Song
- Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Parolini C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem Pharmacol 2025; 235:116844. [PMID: 40044049 DOI: 10.1016/j.bcp.2025.116844] [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: 11/28/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Osteoporosis is the most common skeletal disease worldwide, characterized by low bone mineral density, resulting in weaker bones, and an increased risk of fragility fractures. The maintenance of bone mass relies on the precise balance between bone synthesis and resorption. The close relationship between the immune and skeletal systems, called "osteoimmunology", was coined to identify these overlapping "scientific worlds", and its function resides in the evaluation of the mutual effects of the skeletal and immune systems at the molecular and cellular levels, in both physiological and pathological states. Lipids play an essential role in skeletal metabolism and bone health. Indeed, bone marrow and its skeletal components demand a dramatic amount of daily energy to control hematopoietic turnover, acquire and maintain bone mass, and actively being involved in whole-body metabolism. Statins, the main therapeutic agents in lowering plasma cholesterol levels, are able to promote osteoblastogenesis and inhibit osteoclastogenesis. This review is meant to provide an updated overview of the pathophysiology of bone remodelling cycle, focusing on the interplay between bone, immune system and lipids. Novel therapeutic strategies for the management of osteoporosis are also discussed.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti', via Balzaretti 9 - Università degli Studi di Milano 20133 Milano, Italy.
| |
Collapse
|
6
|
Yashima N, Minamizono W, Matsunaga H, Lyu J, Fujikawa K, Suito H, Okunuki T, Nakai S, Ohsako M. Non-contact electrical stimulation via a Vector-potential transformer promotes bone healing in drill-hole injury model. J Bone Miner Metab 2025:10.1007/s00774-025-01603-0. [PMID: 40301161 DOI: 10.1007/s00774-025-01603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION We investigated the effects of non-contact electrical stimulation via a Vector-potential (VP) transformer, a novel physical therapy device, on bone healing in drill-hole injury models. MATERIALS AND METHODS Six-week-old male Wistar rats, after a one-week acclimation period, were divided into three groups: the control group (CO), the bone injury group (BI), in which a drill-hole injury was created, and the VP stimulation group (VP), which received non-contact electrical stimulation via a VP transformer after bone injury. In the VP group, rats underwent stimulation at 200 kHz for 30 minutes per day, seven days per week. RESULTS The VP group exhibited increased bone formation as early as day 7 post-injury, with significantly higher bone volume than the BI group at all time points (day 7: p = 0.0003; day 14: p = 0.0024; day 21: p = 0.0001). By day 21, the VP group showed lighter toluidine blue staining and reduced biglycan immunoreactivity compared to the BI group. Bone mineral density also increased (p = 0.0008). Osteoblasts in the VP group displayed abundant cytoplasm and a high capacity for osteocalcin synthesis. Additionally, the VP group demonstrated increased expression of Bglap (day 5: p = 0.0068; day 7: p = 0.0096) and Ctsk (day 7: p = 0.0329; day 14: p = 0.0171), along with a higher number of TRAP-positive osteoclasts (day 21: p = 0.0159) compared to the BI group. CONCLUSION Non-contact electrical stimulation via a VP transformer promotes bone healing in drill-hole injury models.
Collapse
Affiliation(s)
- Nao Yashima
- Graduate School of Health and Sports Science, Toyo University, 1-7-11 Akabanedai, Kita-ku, Tokyo, 115-8650, Japan.
| | - Wataru Minamizono
- Graduate School of Human Life Design, Toyo University, 1-7-11 Akabanedai, Kita-ku, Tokyo, 115-8650, Japan
| | - Hiroya Matsunaga
- Graduate School of Health and Sports Science, Toyo University, 1-7-11 Akabanedai, Kita-ku, Tokyo, 115-8650, Japan
| | - Jiazheng Lyu
- Graduate School of Health and Sports Science, Toyo University, 1-7-11 Akabanedai, Kita-ku, Tokyo, 115-8650, Japan
| | - Kaoru Fujikawa
- Department of Oral Anatomy, Showa Medical University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-0064, Japan
| | - Hirai Suito
- Department of Anatomy, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Takumi Okunuki
- Japan Society for the Promotion of Science, Research Organization of Science and Technology, Ritsumeikan University, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Shingo Nakai
- Department of Judo Seifuku and Health Sciences, Tokoha University School of Health Promotional Sciences, 1230 Miyakoda-cho, Hamana-ku, Hamamatsu-shi, Shizuoka, 431-2102, Japan
| | - Masafumi Ohsako
- Department of Health and Sports Science, Toyo University School of Health and Sports Science, 1-7-11 Akabanedai, Kita-ku, Tokyo, 115-8650, Japan
| |
Collapse
|
7
|
Su S, Wang R, Bai J, Gao S, Zhou R, Zhou F. Polydopamine-functionalized acellular fish scale scaffolds for accelerated bone tissue regeneration. RSC Adv 2025; 15:13857-13873. [PMID: 40309128 PMCID: PMC12041853 DOI: 10.1039/d5ra01932j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
The complex microenvironment changes in the bone defect site are the main reason for its refractory treatment, including the significant increase in the level of reactive oxygen species (ROS) and inflammatory dysregulation. There is an urgent need to develop some bioactive materials that can regulate the microenvironment and promote bone regeneration. This study proposed a new strategy for designing bone tissue engineering scaffolds based on fish scales and developed a polydopamine-functionalized acellular fish scale scaffold (PDA-AFS). The results showed that PDA-AFS had excellent mechanical properties, special three-dimensional surface topography, and biodegradability. In vitro results showed that PDA-AFS had good biocompatibility and cell adhesion ability, could effectively reduce ROS levels, and had immunomodulatory activity. More importantly, PDA-AFS can enhance osteogenic differentiation of bone marrow mesenchymal stem cells and promote endogenous bone regeneration in critical-sized calvarial bone defects. In addition, transcriptome analysis also provided clues to its possible osteogenic mechanism. Overall, we provide a new strategy for designing bone tissue engineering scaffolds based on fish scales for bone regeneration treatment of bone defects.
Collapse
Affiliation(s)
- Shilong Su
- Department of Orthopedics, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China +86-01082266699
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China +86-01082266699
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China +86-01082266699
| | - Shan Gao
- Department of Orthopedics, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China +86-01082266699
| | - Rubing Zhou
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University No. 95 Yong'an Road, Xicheng Beijing 100050 China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China
- Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital No. 49 North Garden Road, Haidian Beijing 100191 China +86-01082266699
| |
Collapse
|
8
|
Arvind V, Crosio G, Howell K, Zhang H, Montero A, Huang AH. Functional tendon regeneration is driven by regulatory T cells and IL-33 signaling. SCIENCE ADVANCES 2025; 11:eadn5409. [PMID: 40267206 PMCID: PMC12017337 DOI: 10.1126/sciadv.adn5409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
Tendon injuries heal by scar, leading to poor function. To date, the role of immune cells remains underexplored. Using a neonatal mouse model of functional tendon healing compared to adult scar-mediated healing, we identified a regenerative immune profile that is associated with type 1 inflammation followed by rapid polarization to type 2, driven by macrophages and regulatory T cells (Treg cells). Single-cell and bulk RNA sequencing also revealed neonatal Treg cells with an immunomodulatory signature distinct from adult. Neonatal Treg cell ablation resulted in a dysregulated immune response, failed tenocyte recruitment, and impaired regeneration. Adoptive transfer further confirmed the unique capacity of neonatal Treg cells to rescue functional regeneration. We showed that neonatal Treg cells mitigate interleukin-33 (IL-33) to enable tenocyte recruitment and structural restoration, and that adult IL-33 deletion improves functional healing. Collectively, these findings demonstrate that Treg cells and IL-33 immune dysfunction are critical components of failed tendon healing and identify potential targets to drive tendon regeneration.
Collapse
Affiliation(s)
- Varun Arvind
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Giulia Crosio
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristen Howell
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hui Zhang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Angela Montero
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Alice H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
9
|
Luo G, Zhang Y, Chen P, Wu F, Shi M, Ma Y, Wang X. Tailoring osteo-immunomodulatory micro-environments via a bioactive 3D PLA scaffold to potentiate regenerative healing. Colloids Surf B Biointerfaces 2025; 253:114711. [PMID: 40250081 DOI: 10.1016/j.colsurfb.2025.114711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/31/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
The burden of long bone fractures impacts society profoundly. Polylactic acid (PLA) and PLA-based scaffolds have been widely used for bone tissue engineering due to its biodegradable and non-toxic properties. However, PLA scaffolds possess no biological and immunoregulatory activity. In addition, M2 phenotypic macrophages and M2 macrophage inducer have shown pro-healing effect. Therefore, in this study, we sought to harness the potent osteo-immunomodulatory properties of anti-inflammatory M2 macrophages and encapsulated bioactive interleukin-4 microdroplets (MDs) into the PLA scaffold matrix in order to enhance the biological activity and osteo-immunomodulatory properties of PLA scaffold. The MTT assay, live and dead staining, and phalloidin/DAPI dual staining were used to evaluate biocompatibility in human bone marrow mesenchymal stem cells (hBMSCs). Unstimulated macrophages and inflammatory macrophages were further used to test its immunoregulatory role. The immunoregulatory role of MDs-IL4/PLA hybrid scaffolds on hBMSCs differentiation was further investigated in vitro. The MDs-IL4/PLA hybrid scaffolds showed high biocompatibility in hBMSCs. In addition, MDs-IL4/PLA hybrid scaffolds demonstrated good immunoregulatory role in unstimulated and inflammatory macrophages, as evidenced by marked morphological changes, significantly increased M2 phenotypic markers, and reduction of pro-inflammatory marker expression, etc. Furthermore, our results indicate that the MDs-IL4/PLA hybrid scaffolds could significantly enhance the osteogenic differentiation of hBMSCs via its potential immunomodulatory properties. The subcutaneous implantation of MDs-IL4/PLA scaffold demonstrated significant increases in IL-4, IL-10, CCL2, CCL20, beta-NGF, and TIMP-1 expressions, indicating its notable in vivo immunomodulatory effects. By harnessing the synergistic effects of PLA and MDs-IL4, this novel scaffold holds promise for advancing regenerative medicine approaches, particularly in bone tissue engineering.
Collapse
Affiliation(s)
- Guochen Luo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Maternal and Child Health and Exposure Science, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ping Chen
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Fujun Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Maobiao Shi
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China; Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
10
|
Gupta K, Kaushik N, Sharma V, Singh A. A review on innovations in hydroxyapatite: advancing sustainable and multifunctional dental implants. Odontology 2025:10.1007/s10266-025-01096-3. [PMID: 40208376 DOI: 10.1007/s10266-025-01096-3] [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: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
The increasing relevance of biomaterials in medical curements and the care of aging populations has led to significant advancements in the development and modification of these materials. Hydroxyapatite (HAp), a biocompatible ceramic that mimics the composition of bone mineral, stands out for its remarkable abilities. Its stability in body fluids and ability to integrate with bone without causing toxicity or inflammation made it a prime candidate for biomedical utilization, particularly in odontology and orthopedics. Dental implants, which necessitate a strong interface with the jawbone to effectively support prosthetic devices, are enhanced by coatings of calcium phosphate-based materials like HAp. This enhances osseointegration, ensuring a strong bond and longevity of the implant. HAp may be synthesized both synthetically and from natural sources like mammalian bones, marine shells, and plants, each offering unique trace elements that improve its bioactivity. The synthesis of HAp involves differential technique, including chemical precipitation, and hydrothermal techniques, each impacting the final abilities of the material. The use of natural sources is especially promising, providing a sustainable, cost-effective alternative that retains essential biocompatibility. Hence, this article aims to explore the synthesis, properties, and biomedical applications of hydroxyapatite (HAp), with a special emphasis on its role in improving the performance and durability of dental implants. It also addresses the challenges in manufacturing and biocompatibility, offering insights into future advancements in this field. By addressing current challenges in manufacturing and biocompatibility, HAp paves the way for more effective and long-lasting dental treatments.
Collapse
Affiliation(s)
- Komal Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
- Galgotias College of Pharmacy, Greater Noida, India
| | - Niranjan Kaushik
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India.
| | | | - Amit Singh
- School of Pharmacy, Monad University, Hapur, Uttar Pradesh, India
| |
Collapse
|
11
|
He Y, Lu Y, Li R, Tang Y, Du W, Zhang L, Wu J, Li K, Zhuang W, Lv S, Han Y, Tao B, Deng F, Zhao W, Yu D. CircAars-Engineered ADSCs Facilitate Maxillofacial Bone Defects Repair Via Synergistic Capability of Osteogenic Differentiation, Macrophage Polarization and Angiogenesis. Adv Healthc Mater 2025; 14:e2404501. [PMID: 40035523 PMCID: PMC12004435 DOI: 10.1002/adhm.202404501] [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: 11/13/2024] [Revised: 01/12/2025] [Indexed: 03/05/2025]
Abstract
Adipose-derived stem cells (ADSCs) hold significant promise in bone tissue engineering due to their self-renewal capacity and easy accessibility. However, their limited osteogenic potential remains a critical challenge for clinical application in bone repair. Emerging evidence suggests that circular RNAs (circRNAs) play a key role in regulating stem cell fate and osteogenesis. Despite this, the specific mechanisms by which circRNAs influence ADSCs in the context of bone tissue engineering are largely unexplored. This study introduces a novel strategy utilizing circAars, a specific circRNA, to modify ADSCs, which are then incorporated into gelatin methacryloyl (GelMA) hydrogels for the repair of critical-sized maxillofacial bone defects. The findings reveal that circAars predominantly localizes in the cytoplasm of ADSCs, where it acts as a competitive sponge for miR-128-3p, enhancing the osteogenic differentiation and migration capabilities of ADSCs. Furthermore, circAars-engineered ADSCs facilitate macrophage polarization from the M1 to M2 phenotype and enhance endothelial cell (EC) angiogenic potential through a paracrine mechanism. Additionally, GelMA scaffolds loaded with circAars-engineered ADSCs accelerate the repair of critical-sized maxillofacial bone defects by synergistically promoting osteogenesis, macrophage M2 polarization, and angiogenesis. This approach offers a promising therapeutic strategy for the treatment of critical-sized maxillofacial defects.
Collapse
Affiliation(s)
- Yi He
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yunyang Lu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Runze Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yuquan Tang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Guangzhou Liwan District Stomatological HospitalGuangzhou510080P. R. China
| | - Weidong Du
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Lejia Zhang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Jie Wu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Kechen Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Weijie Zhuang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Shiyu Lv
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yaoling Han
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Bailong Tao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Laboratory Research CenterThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
| | - Feilong Deng
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Wei Zhao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Dongsheng Yu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| |
Collapse
|
12
|
Ning J, Sah RK, Wang J. Coculture of mesenchymal stem cells and macrophage: A narrative review. J Pharmacol Exp Ther 2025; 392:103531. [PMID: 40154096 DOI: 10.1016/j.jpet.2025.103531] [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: 10/31/2024] [Accepted: 02/24/2025] [Indexed: 04/01/2025] Open
Abstract
Stem cell transplantation is a promising treatment for repairing damaged tissues, but challenges like immune rejection and ethical concerns remain. Mesenchymal stem cells (MSCs) offer high differentiation potential and immune regulatory activity, showing promise in treating diseases such as gynecological, neurological, and kidney disorders. With scientific progress, MSC applications are overcoming traditional treatment limitations. In MSCs-macrophage coculture, MSCs transform macrophages into anti-inflammatory M2 macrophages, reducing inflammation, whereas macrophages enhance MSCs osteogenic differentiation. This coculture is vital for immune modulation and tissue repair, with models varying by contact type and dimensional arrangements. Factors such as coculture techniques and cell ratios influence outcomes. Benefits include improved heart function, wound healing, reduced lung inflammation, and accelerated bone repair. Challenges include optimizing coculture conditions. This study reviews the methodologies, factors, and mechanisms of MSC-macrophage coculture, providing a foundation for tissue engineering applications. SIGNIFICANCE STATEMENT: This review underlines the significant role of mesenchymal stem cell-macrophage coculture, providing a foundation for tissue engineering application.
Collapse
Affiliation(s)
- Jun Ning
- Department of General Gynecology II, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, China
| | - Rajiv Kumar Sah
- Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital, Houston, Texas
| | - Jing Wang
- Department of Reproductive Medicine, Department of Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
Zhu Z, Bai Z, Cui Y, Li X, Zhu X. The potential therapeutic effects of Panax notoginseng in osteoporosis: A comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156703. [PMID: 40354676 DOI: 10.1016/j.phymed.2025.156703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Accumulating evidence shows that Panax notoginseng, a well-known medicinal herb, has an ideal effect on prevention and treatment of skeletal diseases. In this study, we reviewed clinical applications of clinical application as well as phytochemistry, pharmacokinetics, pharmacology in improving bone quality and toxicity of Panax notoginseng. PURPOSE Review the phytochemistry, pharmacokinetics, pharmacology involved in the improving bone metabolism and toxicity of Panax notoginseng and evaluate its potential as a traditional Chinese herbal medicine for osteoporosis. METHODS Several databases were consulted, including PubMed, China National Knowledge Infrastructure, National Science and Technology Library and Web of Science. The following words or phrases were used alone or in combinations in the titles and/or abstracts: "","Panax notoginseng", "Sanqi", "osteoporosis", "bone", "osteoblast", "osteoclast", "phytochemistry", "pharmacology" and "pharmacokinetics". Altogether 160 papers were cited. RESULTS 8 clinical trials of Panax notoginseng alone for the treatment of osteoporosis were identified, most of which used traditional Chinese patent medicines to treat osteoporosis fractures. In these clinical trials, Panax notoginseng preparations have achieved relatively good therapeutic effects. However, more rigorous large-scale experiments are expected to prove their efficacy. Phytochemistry study showed that saponins, flavonoids, polysaccharides are the main active ingredients extracted from Panax notoginseng and the transformation of saponins during the processing explains the different effects of raw and cooked Panax notoginseng. The pharmacokinetics data reveals that protopanaxdiol-type (ppd-type) saponins possesses higher bioavailability than protopanaxtriol-type(ppt-type) saponins and ppd-type saponins such as ginsenoside Ra3, Rb1, and Rd can represent suitable pharmacokinetic markers for Panax notoginseng extracts. The data from animal experiments demonstrates that Panax notoginseng can improve bone quality in ovariectomized, diabetic, hyperlipidemia, radiation-induced, and arthritis rats through the regulation of anti-adipogenesis, anti-inflammation, anti-oxidation, angiogenesis and estrogenic effects. In vitro experiments, the activities of improving bone quality of Panax notoginseng and its ingredients may be attributed to the regulation of multiple signaling pathways, including Wnt/β-catenin, BMP/BMP-R, AMPK/mTOR, GPER/PI3K/AKT, etc. Acute and chronic toxicity as well as genotoxicity studies show that Panax notoginseng is well tolerated while long term use may lead to liver and kidney toxicity. CONCLUSIONS Panax notoginseng is a superior medicinal herb that contains multiple active ingredients and could play a potential role in the prevention and treatment of osteoporosis. Further studies should concentrate on developing Panax notoginseng products with higher curative effect and bioavailability.
Collapse
Affiliation(s)
- Zijun Zhu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Zhenyu Bai
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Yan Cui
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - XiaoFeng Zhu
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China; School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
14
|
Negrescu AM, Nistorescu S, Bonciu A, Rusen L, Dumitrescu N, Urzica I, Moldovan A, Hoffmann P, Pircalaboiu GG, Cimpean A, Dinca V. PDMS biointerfaces featuring honeycomb-like well microtextures designed for a pro-healing environment. RSC Adv 2025; 15:9952-9967. [PMID: 40171285 PMCID: PMC11959266 DOI: 10.1039/d5ra00063g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Even today, the reduction of complications following breast implant surgery together with the enhancement of implant integration and performance through the modulation of the foreign body response (FBR), remains a fundamental challenge in the field of plastic surgery. Therefore, tailoring the material's physical characteristics to modulate FBR can represent an effective approach in implantology. While polydimethylsiloxane (PDMS) patterning on 2D substrates is a relatively established and available procedure, micropatterning multiscaled biointerfaces on a controlled large area has been more challenging. Therefore, in the present work, a specific designed honeycomb-like well biointerface was designed and obtained by replication in PDMS at large scale and its effectiveness towards creating a pro-healing environment was investigated. The grayscale masks assisted laser-based 3D texturing method was used for creating the required moulds in Polycarbonate for large area replication. By comparison to the smooth substrate, the honeycomb topography altered the fibroblasts' behaviour in terms of adhesion and morphology and reduced the macrophages' inflammatory response. Additionally, the microstructured surface effectively inhibited macrophage fusion, significantly limiting the colonization of both Gram-positive and Gram-negative microbial strains on the tested surfaces. Overall, this study introduces an innovative approach to mitigate the in vitro FBR to silicone, achieved through the creation of a honeycomb-inspired topography for prosthetic interfaces.
Collapse
Affiliation(s)
- Andreea Mariana Negrescu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- Research Institute of the University of Bucharest (ICUB), University of Bucharest 050657 Bucharest Romania
| | - Simona Nistorescu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Anca Bonciu
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Laurentiu Rusen
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Nicoleta Dumitrescu
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Iuliana Urzica
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Antoniu Moldovan
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| | - Patrick Hoffmann
- Laboratory for Advanced Materials Processing, EMPA, Swiss Federal Institute for Materials Science and Technology Feuerstrasse 39 CH-3602 Thun Swizerland
| | - Gratiela Gradisteanu Pircalaboiu
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
- Polytechnic University of Bucharest Splaiul Independenţei 313 Bucharest 060042 Romania
| | - Anisoara Cimpean
- Faculty of Biology, University of Bucharest Splaiul Independenţei 91-95 050095 Bucharest Romania
| | - Valentina Dinca
- National Institute for Lasers, Plasma, and Radiation Physics 409 Atomistilor Street 077125 Romania
| |
Collapse
|
15
|
Häusner S, Kolb A, Übelmesser K, Hölscher-Doht S, Jordan MC, Jauković A, Berberich-Siebelt F, Spasovski DV, Groll J, Blunk T, Herrmann M. It is not waste if it is therapy: cellular, secretory and functional properties of reamer-irrigator-aspirator (RIA)-derived autologous bone grafts. J Orthop Traumatol 2025; 26:21. [PMID: 40140186 PMCID: PMC11947367 DOI: 10.1186/s10195-025-00835-0] [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: 09/03/2024] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Large bone defects resulting from trauma, disease, or resection often exceed the intrinsic capacity of bones to heal. The current gold standard addressing these defects is autologous bone grafting (ABG). Procedures such as reamer-irrigator-aspirator (RIA) and conventional bone grafting from the iliac crest are widely recognized as highly effective interventions for critical-size bone defects. The early phase of fracture healing is particularly crucial, as it can determine whether a complete bony union occurs, or if delayed healing or non-unions develop. The initial composition of the bone marrow (BM)-rich ABG transplant, with its unique cellular (e.g., leukocytes, monocytes, and granulocytes) and acellular (e.g., growth factors and extracellular proteins) components, plays a key role in this process. However, despite many successful case reports, the role of ABG cells, growth factors, and their precise contributions to bone healing remain largely elusive. MATERIALS AND METHODS We characterized the native cellularity of both solid and liquid RIA-derived ABG by analyzing primary, minimally manipulated populations of monocytes, macrophages, and T cells, as well as hematopoietic, endothelial, and mesenchymal progenitor cells by flow cytometry. Growth factor and cytokine contents were assessed through antibody arrays. Possible functional and immunomodulatory properties of RIA liquid were evaluated in functional in vitro assays. RESULTS Growth factor and protein arrays revealed a plethora of soluble factors that can be linked to specific immunomodulatory and angiogenic properties, which were evaluated for their potency using functional in vitro assays. We could demonstrate a strong M2-macrophage phenotype inducing the effect of RIA liquid on macrophages. Additionally, we observed an increase in anti-inflammatory T cell subsets generated from peripheral blood mononuclear cells and BM mononuclear cells upon stimulation with RIA liquid . Finally, in vitro endothelial tube formation assays revealed highly significant angiogenic properties of RIA liquid, even at further dilutions. CONCLUSION The cytokine and protein content of RIA liquid exhibits potent immunomodulatory and angiogenic properties. These findings suggest significant therapeutic potential for RIA liquid in modulating immune responses and promoting angiogenesis. Anti-inflammatory and angiogenic properties demonstrated in this study might also help to further define and understand its particular mode of action while also providing explanations to the excellent bone-healing properties of ABG in general. LEVEL OF EVIDENCE Case-series (Level 4).
Collapse
Affiliation(s)
- S Häusner
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Bernhard-Heine-Center for Locomotion Research, Chair of Orthopedics, University of Würzburg, Brettreichstr. 11, 97074, Würzburg, Germany.
| | - A Kolb
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - K Übelmesser
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - S Hölscher-Doht
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery (Surgery II), University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - M C Jordan
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - A Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, P.O.B. 102, 11129, Belgrade, Serbia
| | - F Berberich-Siebelt
- Institute of Pathology, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - D V Spasovski
- Institute for Orthopedic Surgery (Banjica), University of Belgrade, Milhaila Avramovica 28, Belgrade, Serbia
| | - J Groll
- Department for Functional Materials in Medicine and Dentistry (FMZ), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - T Blunk
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery (Surgery II), University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - M Herrmann
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Bernhard-Heine-Center for Locomotion Research, Chair of Orthopedics, University of Würzburg, Brettreichstr. 11, 97074, Würzburg, Germany.
| |
Collapse
|
16
|
Xu K, Wu K, Chen L, Zhao Y, Li H, Lin N, Ye Z, Xu J, Huang D, Huang X. Selective promotion of sensory innervation-mediated immunoregulation for tissue repair. SCIENCE ADVANCES 2025; 11:eads9581. [PMID: 40117376 PMCID: PMC11927663 DOI: 10.1126/sciadv.ads9581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025]
Abstract
Sensory innervation triggers the regenerative response after injury. However, dysfunction and impairment of sensory nerves, accompanied by excessive inflammation impede tissue regeneration. Consequently, specific induction of sensory innervation to mediate immunoregulation becomes a promising therapeutic approach. Herein, we developed a cell/drug-free strategy to selectively boost endogenous sensory innervation to harness immune responses for promoting tissue rehabilitation. Specifically, a dual-functional phage was constructed with a sensory nerve-homing peptide and a β-subunit of nerve growth factor (β-NGF)-binding peptide. These double-displayed phages captured endogenic β-NGF and localized to sensory nerves to promote sensory innervation. Furthermore, regarding bone regeneration, phage-loaded hydrogels achieved rapid sensory nerve ingrowth in bone defect areas. Mechanistically, sensory neurotization facilitated M2 polarization of macrophages through the Sema3A/XIAP/PAX6 pathway, thus decreasing the M1/M2 ratio to induce the dissipation of local inflammation. Collectively, these findings highlight the essential role of sensory innervation in manipulating inflammation and provide a conceptual framework based on neuroimmune interactions for promoting tissue regeneration.
Collapse
Affiliation(s)
- Kaicheng Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Kaile Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Liang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yubin Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Nong Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Donghua Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xin Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Shi A, Shi Y, Li J, Ye M, Ma X, Peng Y, Gai K, Chen J. Advancements in 3D gel culture systems for enhanced angiogenesis in bone tissue engineering. J Mater Chem B 2025; 13:3516-3527. [PMID: 39998426 DOI: 10.1039/d4tb01139b] [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: 02/26/2025]
Abstract
Angiogenesis-osteogenesis coupling is a crucial process in bone tissue engineering, requiring a suitable material structure for vessel growth. Recently, the 3D culture system has gained significant attention due to its benefits in cell growth, proliferation and tissue regeneration. Its most notable advantage is its ECM-like function, which supports endothelial cell adhesion and facilitates the formation of vascular-like networks-crucial for angiogenesis-osteogenesis coupling. Hydrogels, with their highly hydrophilic polymer network resembling the extracellular matrix, make the 3D gel culture system an ideal approach for angiogenesis due to its cellular integrity and adjustable properties. This article reviews the current use of 3D gel culture systems in bone tissue engineering, covering substrates, characteristics and processing technologies, thereby offering readers profound insights into these systems.
Collapse
Affiliation(s)
- Aijing Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yixin Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Jie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Minghan Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xiaoqing Ma
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yuke Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Kuo Gai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China.
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
18
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
Al-Shamaa RM, Al-Askary RA. Remineralizing capacity of zinc oxide eugenol sealer following the addition of nanohydroxyapatite-tyrosine amino acid: An in vivo animal study. J Oral Biosci 2025; 67:100567. [PMID: 39419338 DOI: 10.1016/j.job.2024.09.006] [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: 03/28/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Although several sealers have been developed, the zinc oxide eugenol (ZOE) sealer is still used in many private dental clinics. This study aimed to compare the biocompatibility and remineralizing capacity οf ZOE sealer with the addition of nanohydroxyapatite-tyrosine amino acid. METHODS This study was conducted on Twenty rabbits. The rabbits were divided into four groups based on the test observation period (3, 7, 21, and 28 days) following surgical implantation. General anesthesia was administered to each rabbit, and a subcutaneous incision of approximately 1 ± 0.5 cm was made along the symphyseal area of the mandible of each rabbit. Four bone cavities were generated in the interdental space of the lower jaw between the central and molar teeth of each rabbit, with one longitudinal subcutaneous incision. The ZOE sealers were mixed according to the manufacturer's guidelines and directly inserted within the cavities generated at the end of each test period. The animals were sacrificed, and bone biopsy was carried out at the site of testing. The biopsy samples were obtained and subjected to histological analysis using a low-power light microscope (Olympus C ⅹ 21, Japan) and immunohistochemistry using Ki67 antibody. RESULTS The collected information was examined by parametric statistical tests using SPSS software version ''22." One-way ANOVA and post-hoc Duncan's tests were used to measure the significance among various groups, with statistical significance set, when "P ≤ 0.01". CONCLUSION The 20% mixed endodontic sealer displayed excellent outcomes compared to other experimental groups, as identified by higher new bone formation at every evaluation period.
Collapse
Affiliation(s)
- Rasha M Al-Shamaa
- Department of Conservative Dentistry, College of Dentistry, Mosul University, 41001, Iraq.
| | - Raghad A Al-Askary
- Department of Conservative Dentistry, College of Dentistry, Mosul University, 41001, Iraq.
| |
Collapse
|
20
|
Zhao J, Sarkar N, Ren Y, Pathak AP, Grayson WL. Engineering next-generation oxygen-generating scaffolds to enhance bone regeneration. Trends Biotechnol 2025; 43:540-554. [PMID: 39343620 PMCID: PMC11867879 DOI: 10.1016/j.tibtech.2024.09.006] [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: 03/14/2024] [Revised: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
In bone, an adequate oxygen (O2) supply is crucial during development, homeostasis, and healing. Oxygen-generating scaffolds (OGS) have demonstrated significant potential to enhance bone regeneration. However, the complexity of O2 delivery and signaling in vivo makes it challenging to tailor the design of OGS to precisely meet this biological requirement. We review recent advances in OGS and analyze persisting engineering and translational hurdles. We also discuss the potential of computational and machine learning (ML) models to facilitate the integration of novel imaging data with biological readouts and advanced biomanufacturing technologies. By elucidating how to tackle current challenges using cutting-edge technologies, we provide insights for transitioning from traditional to next-generation OGS to improve bone regeneration in patients.
Collapse
Affiliation(s)
- Jingtong Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Naboneeta Sarkar
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
21
|
Saiz AM, Rahmati M, Gresham RCH, Baldini TD, Burgan J, Lee MA, Osipov B, Christiansen BA, Khassawna TE, Wieland DCF, Marinho AL, Blanchet C, Czachor M, Working ZM, Bahney CS, Leach JK. Polytrauma impairs fracture healing accompanied by increased persistence of innate inflammatory stimuli and reduced adaptive response. J Orthop Res 2025; 43:603-616. [PMID: 39550711 PMCID: PMC11806648 DOI: 10.1002/jor.26015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
The field of bone regeneration has primarily focused on investigating fracture healing and nonunion in isolated musculoskeletal injuries. Compared to isolated fractures, which frequently heal well, fractures in patients with multiple bodily injuries (polytrauma) may exhibit impaired healing. While some papers have reported the overall cytokine response to polytrauma conditions, significant gaps in our understanding remain in how fractures heal differently in polytrauma patients. We aimed to characterize fracture healing and the temporal local and systemic immune responses to polytrauma in a murine model of polytrauma composed of a femur fracture combined with isolated chest trauma. We collected serum, bone marrow from the uninjured limb, femur fracture tissue, and lung tissue over 3 weeks to study the local and systemic immune responses and cytokine expression after injury. Immune cell distribution was assessed by flow cytometry. Fracture healing was characterized using microcomputed tomography (microCT), histological staining, immunohistochemistry, mechanical testing, and small angle X-ray scattering. We detected more innate immune cells in the polytrauma group, both locally at the fracture site and systemically, compared to other groups. The percentage of B and T cells was dramatically reduced in the polytrauma group 6 h after injury and remained low throughout the study duration. Fracture healing in the polytrauma group was impaired, evidenced by the formation of a poorly mineralized and dysregulated fracture callus. Our data confirm the early, dysregulated inflammatory state in polytrauma that correlates with disorganized and impaired fracture healing.
Collapse
Affiliation(s)
| | - Maryam Rahmati
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | | | - Tony Daniel Baldini
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
- California Northstate University College of MedicineSacramentoCaliforniaUSA
| | - Jane Burgan
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
- Stony Brook Renaissance School of MedicineStony BrookNew YorkUSA
| | - Mark A. Lee
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | - Benjamin Osipov
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| | | | - Thaqif El Khassawna
- Experimental Trauma SurgeryJustus‐Liebig University GiessenGiessenGermany
- Faculty of Health SciencesUniversity of Applied SciencesGiessenGermany
| | | | - André Lopes Marinho
- Institute of Metallic Biomaterials, Helmholtz Zentrum HereonGeesthachtGermany
| | | | - Molly Czachor
- Steadman Phillippon Research InstituteVailColoradoUSA
| | | | - Chelsea S. Bahney
- Steadman Phillippon Research InstituteVailColoradoUSA
- University of CaliforniaSan FranciscoCaliforniaUSA
| | - J. Kent Leach
- Department of Orthopaedic SurgeryUC Davis HealthSacramentoCaliforniaUSA
| |
Collapse
|
22
|
Dong W, Yang C, Guo D, Jia M, Wang Y, Wang J. PTX3-assembled pericellular hyaluronan matrix enhances endochondral ossification during fracture healing and heterotopic ossification. Bone 2025; 192:117385. [PMID: 39732447 DOI: 10.1016/j.bone.2024.117385] [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: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Endochondral ossification (EO) is a pivotal process during fracture healing and traumatic heterotopic ossification (HO), involving the cartilaginous matrix synthesis and mineralization. Unlike the extracellular matrix, the hyaluronan (HA)-rich pericellular matrix (PCM) directly envelops chondrocytes, serving as the frontline for extracellular signal reception and undergoing dynamic remodeling. Pentraxin 3 (PTX3), a secreted glycoprotein, facilitates HA matrix assembly and remodeling. However, it remains unclear whether PTX3 affects EO by regulating HA-rich PCM assembly of chondrocytes, thereby impacting fracture healing and traumatic HO. This study demonstrates that PTX3 deficiency impairs fracture healing and inhibits traumatic HO, but dose not affect growth plate development in mice. PTX3 expression is up-regulated during chondrocyte matrix synthesis and maturation and is localized in the HA-rich PCM. PTX3 promotes the assembly of HA-rich PCM in a serum- and TSG6-dependent manner, fostering CD44 receptor clustering, activating the FAK/AKT signaling pathway, and promoting chondrocyte matrix synthesis and maturation. Local injection of PTX3/TSG6 matrix protein mixture effectively promotes fracture healing in mice. In conclusion, PTX3-assembled HA-rich PCM promotes chondrocyte matrix synthesis and maturation via CD44/FAK/AKT signaling. This mechanism facilitates EO during fracture healing and traumatic HO in mice.
Collapse
Affiliation(s)
- Wei Dong
- 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 430079, China
| | - Chang Yang
- 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 430079, China
| | - Donghua Guo
- 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 430079, China
| | - Meie Jia
- 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 430079, China
| | - Yan 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 430079, China
| | - Jiawei 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 430079, China.
| |
Collapse
|
23
|
Wang H, Li Y, Li H, Yan X, Jiang Z, Feng L, Hu W, Fan Y, Lin S, Li G. T cell related osteoimmunology in fracture healing: Potential targets for augmenting bone regeneration. J Orthop Translat 2025; 51:82-93. [PMID: 39991456 PMCID: PMC11847249 DOI: 10.1016/j.jot.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 02/25/2025] Open
Abstract
UNLABELLED Last decade has witnessed increasing evidence which highlights the roles of immune cells in bone regeneration. Numerous immune cell types, including macrophages, T cells, and neutrophils are involved in fracture healing by orchestrating a series of events that modulate bone formation and remodeling. In this review, the role of T cell immunity in fracture healing has been summarized, and the modulatory effects of T cell immunity in inflammation, bone formation and remodeling have been highlighted. The review also summarizes the specific roles of different T cell subsets, including CD4+ T cells, CD8+ T cells, regulatory T cells, T helper 17 cells, and γδ T cells in modulating fracture healing. The current therapeutics targeting T cell immunity to enhance fracture healing have also been reviewed, aiming to provide insights from a translational standpoint. Overall, this work discusses recent advances and challenges in the interdisciplinary research field of T cell related osteoimmunology and its implications in fracture healing. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Delayed unions or non-unions of bone fractures remain a challenge in clinical practice. Developing a deep understanding of the roles of immune cells, including T cells, in fracture healing will facilitate the advancement of novel therapeutics of fracture nonunion. This review summarizes the current understanding of different T cell subsets involved in various phases of fracture healing, providing insights for targeting T cells as an alternative strategy to enhance bone regeneration.
Collapse
Affiliation(s)
- Haixing Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yashi Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haoxin Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Yan
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhaowei Jiang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Wenhui Hu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yinuo Fan
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
24
|
Perepletchikova D, Malashicheva A. Communication between endothelial cells and osteoblasts in regulation of bone homeostasis: Notch players. Stem Cell Res Ther 2025; 16:56. [PMID: 39920854 PMCID: PMC11806792 DOI: 10.1186/s13287-025-04176-x] [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: 07/25/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Endothelial cells coat blood vessels and release molecular signals to affect the fate of other cells. Endothelial cells can adjust their behavior in response to the changing microenvironmental conditions. During bone regeneration, bone tissue cells release factors that promote blood vessel growth. Notch is a key signaling that regulates cell fate decisions in many tissues and plays an important role in bone tissue development and homeostasis. Understanding the interplay between angiogenesis and osteogenesis is currently a focus of research efforts in order to facilitate and improve osteogenesis when needed. Our review explores the cellular and molecular mechanisms including Notch-dependent endothelial-MSC communication that drive osteogenesis-angiogenesis processes and their effects on bone remodeling and repair.
Collapse
Affiliation(s)
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia, 194064.
| |
Collapse
|
25
|
Liu L, Chen H, Zhao X, Han Q, Xu Y, Liu Y, Zhang A, Li Y, Zhang W, Chen B, Wang J. Advances in the application and research of biomaterials in promoting bone repair and regeneration through immune modulation. Mater Today Bio 2025; 30:101410. [PMID: 39811613 PMCID: PMC11731593 DOI: 10.1016/j.mtbio.2024.101410] [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: 07/06/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
With the ongoing development of osteoimmunology, increasing evidence indicates that the local immune microenvironment plays a critical role in various stages of bone formation. Consequently, modulating the immune inflammatory response triggered by biomaterials to foster a more favorable immune microenvironment for bone regeneration has emerged as a novel strategy in bone tissue engineering. This review first examines the roles of various immune cells in bone tissue injury and repair. Then, the contributions of different biomaterials, including metals, bioceramics, and polymers, in promoting osteogenesis through immune regulation, as well as their future development directions, are discussed. Finally, various design strategies, such as modifying the physicochemical properties of biomaterials and integrating bioactive substances, to optimize material design and create an immune environment conducive to bone formation, are explored. In summary, this review comprehensively covers strategies and approaches for promoting bone tissue regeneration through immune modulation. It offers a thorough understanding of current research trends in biomaterial-based immune regulation, serving as a theoretical reference for the further development and clinical application of biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Li Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xue Zhao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Qing Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongjun Xu
- Department of Orthopedics Surgery, Wangqing County People's Hospital, Yanbian, 133000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yongyue Li
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Weilong Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Bingpeng Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China
| |
Collapse
|
26
|
Zhang Y, Zhou C, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Dual release scaffolds as a promising strategy for enhancing bone regeneration: an updated review. Nanomedicine (Lond) 2025; 20:371-388. [PMID: 39891431 PMCID: PMC11812394 DOI: 10.1080/17435889.2025.2457317] [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: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Advancements in tissue regeneration, particularly bone regeneration is key area of research due to potential of novel therapeutic approaches. Efforts to reduce reliance on autologous and allogeneic bone grafts have led to the development of biomaterials that promote synchronized and controlled bone healing. However, the use of growth factors is limited by their short half-life, slow tissue penetration, large molecular size and potential toxicity. These factors suggest that traditional delivery methods may be inadequate hence, to address these challenges, new strategies are being explored. These novel approaches include the use of bioactive substances within advanced delivery systems that enable precise spatiotemporal control. Dual-release composite scaffolds offer a promising solution by reducing the need for multiple surgical interventions and simplifying the treatment process. These scaffolds allow for sustained and controlled drug release, enhancing bone repair while minimizing the drawbacks of conventional methods. This review explores various dual-drug release systems, discussing their modes of action, types of drugs used and release mechanisms to improve bone regeneration.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, Zhejiang, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
27
|
Dou Y, Zhang Y, Liu Y, Sun X, Liu X, Li B, Yang Q. Role of macrophage in intervertebral disc degeneration. Bone Res 2025; 13:15. [PMID: 39848963 PMCID: PMC11758090 DOI: 10.1038/s41413-024-00397-7] [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: 07/01/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Intervertebral disc degeneration is a degenerative disease where inflammation and immune responses play significant roles. Macrophages, as key immune cells, critically regulate inflammation through polarization into different phenotypes. In recent years, the role of macrophages in inflammation-related degenerative diseases, such as intervertebral disc degeneration, has been increasingly recognized. Macrophages construct the inflammatory microenvironment of the intervertebral disc and are involved in regulating intervertebral disc cell activities, extracellular matrix metabolism, intervertebral disc vascularization, and innervation, profoundly influencing the progression of disc degeneration. To gain a deeper understanding of the inflammatory microenvironment of intervertebral disc degeneration, this review will summarize the role of macrophages in the pathological process of intervertebral disc degeneration, analyze the regulatory mechanisms involving macrophages, and review therapeutic strategies targeting macrophage modulation for the treatment of intervertebral disc degeneration. These insights will be valuable for the treatment and research directions of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215007, China.
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
28
|
Fonseca AC, Colavite PM, Azevedo MDCS, Passadori DC, Melchiades JL, Ortiz RC, Rodini CO, Trombone APF, Garlet GP. Inhibition of MEK1/2 Signaling Pathway Limits M2 Macrophage Polarization and Interferes in the Dental Socket Repair Process in Mice. BIOLOGY 2025; 14:107. [PMID: 40001875 PMCID: PMC11851886 DOI: 10.3390/biology14020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
Dental socket repair theoretically involves a constructive inflammatory immune response, which evolves from an initial M1 prevalence to a subsequent M2 dominance. In this scenario, the MEK1/2 signaling pathway is allegedly involved in M2 polarization. This study aimed to evaluate the impact of MEK1/2 pharmacological inhibition in the local host response and repair outcome. C57Bl/6-WT 8-week-old male mice were submitted to the extraction of the right upper incisor and treated (or not, control group) with MEK1/2 inhibitor PD0325901 (10 mg/kg/24 h/IP, MEK1/2i group) and analyzed at 0, 3, 7, and 14 days using microcomputed tomography, histomorphometry, birefringence, immunohistochemistry, and PCR array analysis. The results demonstrate that MEK1/2 inhibition limits the development of M2 response over time, being associated with lower expression of M2, MSCs, and bone markers, lower levels of growth and osteogenic factors, along with a higher expression of iNOS, IL-1b, IL-6, and TNF-α, as well inflammatory chemokines, indicating a predominantly M1 pro-inflammatory environment. This modulation of local inflammatory immune response is associated with impaired bone formation as demonstrated by microtomographic and histomorphometric data. The results show that MEK1/2 inhibition delays bone repair after tooth extraction, supporting the concept that M2 macrophages are essential elements for host response regulation and proper repair.
Collapse
Affiliation(s)
- Angélica Cristina Fonseca
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Priscila Maria Colavite
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Michelle de Campos Soriani Azevedo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Daniela Carignatto Passadori
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Jessica Lima Melchiades
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Rafael Carneiro Ortiz
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Camila Oliveira Rodini
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| | - Ana Paula Favaro Trombone
- Department of Health Sciences, Centro Universitário Sagrado Coração—UNISAGRADO, Bauru 17011-160, SP, Brazil;
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisola, 9-75, Bauru 17012-901, SP, Brazil; (A.C.F.); (P.M.C.); (M.d.C.S.A.); (D.C.P.); (J.L.M.); (R.C.O.); (C.O.R.)
| |
Collapse
|
29
|
Griffith JW, Luster AD. No bones about it: regulatory T cells promote fracture healing. J Clin Invest 2025; 135:e188368. [PMID: 39817452 PMCID: PMC11735088 DOI: 10.1172/jci188368] [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] [Indexed: 01/18/2025] Open
Abstract
Regulatory T cells (Tregs) are increasingly being recognized for their role in promoting tissue repair. In this issue of the JCI, Chen et al. found that Tregs at the site of bone injury contribute to bone repair. The CCL1/CCR8 chemokine system promoted the accumulation of Tregs at the site of bone injury, where Tregs supported skeletal stem cell (SSC) accumulation and osteogenic differentiation. CCL1 increased the transcription factor basic leucine zipper ATF-like transcription factor (BATF) in CCR8+ Tregs, which induced the secretion of progranulin that promoted SSC osteogenic function and new bone formation. This study highlights the ever-expanding role of Tregs in tissue repair by demonstrating their ability to expand stem cells at a site of injury.
Collapse
Affiliation(s)
- Jason W. Griffith
- Center for Immunology and Inflammatory Diseases
- Division of Pulmonary & Critical Care Medicine, and
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases
- Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Lang A, Collins JM, Nijsure MP, Belali S, Khan MP, Moharrer Y, Schipani E, Yien YY, Fan Y, Gelinsky M, Vinogradov SA, Koch C, Boerckel JD. Local erythropoiesis directs oxygen availability in bone fracture repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632440. [PMID: 39829797 PMCID: PMC11741344 DOI: 10.1101/2025.01.10.632440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Bone fracture ruptures blood vessels and disrupts the bone marrow, the site of new red blood cell production (erythropoiesis). Current dogma holds that bone fracture causes severe hypoxia at the fracture site, due to vascular rupture, and that this hypoxia must be overcome for regeneration. Here, we show that the early fracture site is not hypoxic, but instead exhibits high oxygen tension (> 55 mmHg, or 8%), similar to the red blood cell reservoir, the spleen. This elevated oxygen stems not from angiogenesis but from activated erythropoiesis in the adjacent bone marrow. Fracture-activated erythroid progenitor cells concentrate oxygen through haemoglobin formation. Blocking transferrin receptor 1 (CD71)-mediated iron uptake prevents oxygen binding by these cells, induces fracture site hypoxia, and enhances bone repair through increased angiogenesis and osteogenesis. These findings upend our current understanding of the early phase of bone fracture repair, provide a mechanism for high oxygen tension in the bone marrow after injury, and reveal an unexpected and targetable role of erythroid progenitors in fracture repair.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Joseph M. Collins
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P. Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Simin Belali
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Yvette Y. Yien
- Division of Hematology/Oncology, Department of Medicine and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sergei A. Vinogradov
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Cameron Koch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Shi J, Liu Z, Ren X, Wang W, Zhang H, Wang Y, Liu M, Yao Q, Wu W. Bioinspired adhesive polydopamine-metal-organic framework functionalized 3D customized scaffolds with enhanced angiogenesis, immunomodulation, and osteogenesis for orbital bone reconstruction. Int J Biol Macromol 2025; 284:137968. [PMID: 39581418 DOI: 10.1016/j.ijbiomac.2024.137968] [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: 07/18/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Critical-sized orbital bone defects can lead to significant maxillofacial deformities and even eye movement disorders. The challenges associated with these defects, including their complicated structure, inadequate blood supply, and limited availability of progenitor cells that hinder successful repair. To overcome these issues, we developed a novel approach using computer numerical control (CNC) material reduction manufacturing technology to produce a customized polyetheretherketone (PEEK) scaffold that conforms to the specific shape of orbital bone defects. Deferoxamine (DFO) was in situ encapsulated into polydopamine-hybridized zeolitic imidazolate framework-8 (pZIF8-DFO) nanoparticles, which was subsequently adhered to the sulfonated PEEK (sPEEK) scaffold through polydopamine modification. This functionalization enhanced drug loading efficiency and imparted anti-inflammatory properties to the nanoparticle system. Our in vitro findings demonstrated that the sustained release of DFO from the sPEEK/pZIF8-DFO scaffolds extended over 14 days and significantly promoted angiogenesis and progenitor cell recruitment, as evidenced by increased expression of HIF-1α, VEGF, and SDF-1α expression in human umbilical vein endothelial cells (HUVECs). Moreover, sPEEK/pZIF8-DFO scaffolds exhibited superior immunomodulation and osteogenic differentiation capabilities on Raw 264.7 cells and rabbit bone marrow mesenchymal stem cells (rBMSCs), respectively. Most notably, our in vivo rabbit orbital bone defects revealed that sPEEK/pZIF8-DFO scaffolds resulted in a greater volume of new bone formation than on sPEEK and sPEEK/pZIF8 scaffolds, with partial bone connection to the sPEEK/pZIF8-DFO scaffolds. In summary, we develop a novel PEEK scaffold that combines enhanced angiogenesis, stem cell recruitment, immunomodulation, and osteogenic differentiation, showcasing its promising potential for orbital bone reconstruction.
Collapse
Affiliation(s)
- Jieliang Shi
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Zhirong Liu
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Xiaobin Ren
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Wei Wang
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Haojie Zhang
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Yuanli Wang
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Mingyue Liu
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China
| | - Qingqing Yao
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China.
| | - Wencan Wu
- National Clinical Research Center for Ocular Diseases, National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou 325027, China.
| |
Collapse
|
32
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [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: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
33
|
Li XR, Deng QS, He SH, Liu PL, Gao Y, Wei ZY, Zhang CR, Wang F, Zhu TH, Dawes H, Rui BY, Tao SC, Guo SC. 3D cryo-printed hierarchical porous scaffolds provide immobilization of surface-functionalized sleep-inspired small extracellular vesicles: synergistic therapeutic strategies for vascularized bone regeneration based on macrophage phenotype modulation and angiogenesis-osteogenesis coupling. J Nanobiotechnology 2024; 22:764. [PMID: 39695679 DOI: 10.1186/s12951-024-02977-5] [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: 11/02/2023] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Bone defect healing is a multi-factorial process involving the inflammatory microenvironment, bone regeneration and the formation of blood vessels, and remains a great challenge in clinical practice. Combined use of three-dimensional (3D)-printed scaffolds and bioactive factors is an emerging strategy for the treatment of bone defects. Scaffolds can be printed using 3D cryogenic printing technology to create a microarchitecture similar to trabecular bone. Melatonin (MT) has attracted attention in recent years as an excellent factor for promoting cell viability and tissue repair. In this study, porous scaffolds were prepared by cryogenic printing with poly(lactic-co-glycolic acid) and ultralong hydroxyapatite nanowires. The hierarchical pore size distribution of the scaffolds was evaluated by scanning electron microscopy (SEM) and micro-computed tomography (micro-CT). Sleep-inspired small extracellular vesicles (MT-sEVs) were then obtained from MT-stimulated cells and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol)-inorganic pyrophosphate (DSPE-PEG-PPi) was used to modify the membrane of MT-sEVs to obtain PPi-MT-sEVs. RNA sequencing was performed to explore the potential mechanisms. The results demonstrated that PPi-MT-sEVs not only enhanced cell proliferation, migration and angiogenesis, but also regulated the osteogenic/adipogenic fate determination and M1/M2 macrophage polarization switch in vitro. PPi-MT-sEVs were used to coat scaffolds, enabled by the capacity of PPi to bind to hydroxyapatite, and computational simulations were used to analyze the interfacial bonding of PPi and hydroxyapatite. The macrophage phenotype-modulating and osteogenesis-angiogenesis coupling effects were evaluated in vivo. In summary, this study suggests that the combination of hierarchical porous scaffolds and PPi-MT-sEVs could be a promising candidate for the clinical treatment of bone defects.
Collapse
Affiliation(s)
- Xu-Ran Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Qing-Song Deng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Shu-Hang He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Po-Lin Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yuan Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zhan-Ying Wei
- Shanghai Clinical Research Centre of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chang-Ru Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Clinical and Translational Research Center for 3D Printing Technology, Medical 3D Printing Innovation Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Fei Wang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
| | - Tong-He Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
| | - Helen Dawes
- Faculty of Health and Life Science, Oxford Brookes University, Headington Road, Oxford, OX3 0BP, UK
- NIHR Oxford Health Biomedical Research Centre, Oxford, OX3 7JX, UK
- College of Medicine and Health, St Lukes Campus, University of Exeter, Heavitree Road, Exeter, EX1 2LU, UK
| | - Bi-Yu Rui
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China.
| | - Shang-Chun Guo
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai, 200025, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
34
|
Yang L, Xiao L, Gao W, Huang X, Wei F, Zhang Q, Xiao Y. Macrophages at Low-Inflammatory Status Improved Osteogenesis via Autophagy Regulation. Tissue Eng Part A 2024; 30:e766-e779. [PMID: 33678009 DOI: 10.1089/ten.tea.2021.0015] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence indicates that the interaction between immune and skeletal systems is vital in bone homeostasis. However, the detailed mechanisms between macrophage polarization and osteogenic differentiation of mesenchymal stromal cells (bone marrow-derived stromal cells [BMSCs]) remain largely unknown. We observed enhanced macrophage infiltration along with bone formation in vivo, which showed a transition from early-stage M1 phenotype to later stage M2 phenotype, cells at the transitional stage expressed both M1 and M2 markers that actively participated in osteogenesis, which was mimicked by stimulating macrophages with lower inflammatory stimulus (compared with typical M1). Using conditioned medium (CM) from M0, typical M1, low-inflammatory M1 (M1semi), and M2 macrophages, it was found that BMSCs treated with M1semi CM showed significantly induced migration, osteogenic differentiation, and mineralization, compared with others. Along with the induced osteogenesis, the autophagy level was the highest in M1semi CM-treated BMSCs, which was responsible for BMSC migration and osteogenic differentiation, as autophagy interruption significantly abolished this effect. This study indicated that low-inflammatory macrophages could activate autophagy in BMSCs to improve osteogenesis.
Collapse
Affiliation(s)
- Lan Yang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Lan Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Wendong Gao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Xin Huang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Fei Wei
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Qing Zhang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yin Xiao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
35
|
Kannan R, Koh AJ, Kent RN, Bhutada K, Wasi F, Wagner L, Kozloff K, Baker BM, Roca H, McCauley LK. CCL2/CCR2 Signalling in Mesenchymal Stem/Progenitor Cell Recruitment and Fracture Healing in Mice. J Cell Mol Med 2024; 28:e70300. [PMID: 39721002 DOI: 10.1111/jcmm.70300] [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: 08/27/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024] Open
Abstract
Macrophage efferocytosis (clearance of apoptotic cells) is crucial for tissue homeostasis and wound repair, where macrophages secrete factors that promote resolution of inflammation and regenerative signalling. This study examined the role of efferocytic macrophage-associated CCL2 secretion, its influence on mesenchymal stem/progenitor cell (MSPC) chemotaxis, and in vivo cell recruitment using Ccr2-/- (KO) mice with disrupted CCL2 receptor signalling in two regenerative models: ossicle implants and ulnar stress fractures. Single cell RNA sequencing and PCR validation indicated that efferocytosis of various apoptotic cells at bone injury sites (osteoblasts, pre-osteoblasts, MSPC) upregulated CCL2. CCL2 gradients enhanced MSPC migration through type I collagen matrices. In vivo, MSPC (LepR+) infiltration was significantly reduced while macrophage (F4/80+) infiltration increased in KO ossicle implants versus WT. In ulnar stress fractures, micro-CT revealed increased mineralized callus incidence in CCR2 KO male mice 5 days post injury (dpi) versus WT. By 7-dpi callus fractional bone volume, trabecular thickness, and bone mineral density were increased versus WT. Immunohistochemistry of mice 5-dpi confirmed an increase in callus area (including soft tissue); however, the percent of osteoprogenitors (%Osx+) within the callus was not different. These findings suggest that CCL2 differentially impacts MSPC recruitment depending on bone wound healing model.
Collapse
Affiliation(s)
- Rahasudha Kannan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Kaira Bhutada
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Fatima Wasi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Leon Wagner
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Kenneth Kozloff
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Hernan Roca
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
Rehak L, Giurato L, Monami M, Meloni M, Scatena A, Panunzi A, Manti GM, Caravaggi CMF, Uccioli L. The Immune-Centric Revolution Translated into Clinical Application: Peripheral Blood Mononuclear Cell (PBMNC) Therapy in Diabetic Patients with No-Option Critical Limb-Threatening Ischemia (NO-CLTI)-Rationale and Meta-Analysis of Observational Studies. J Clin Med 2024; 13:7230. [PMID: 39685690 DOI: 10.3390/jcm13237230] [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: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic limb-threatening ischemia (CLTI), the most advanced form of peripheral arterial disease (PAD), is the comorbidity primarily responsible for major lower-limb amputations, particularly for diabetic patients. Autologous cell therapy has been the focus of efforts over the past 20 years to create non-interventional therapeutic options for no-option CLTI to improve limb perfusion and wound healing. Among the different available techniques, peripheral blood mononuclear cells (PBMNC) appear to be the most promising autologous cell therapy due to physio-pathological considerations and clinical evidence, which will be discussed in this review. A meta-analysis of six clinical studies, including 256 diabetic patients treated with naive, fresh PBMNC produced via a selective filtration point-of-care device, was conducted. PBMNC was associated with a mean yearly amputation rate of 15.7%, a mean healing rate of 62%, and a time to healing of 208.6 ± 136.5 days. Moreover, an increase in TcPO2 and a reduction in pain were observed. All-cause mortality, with a mean rate of 22.2% and a yearly mortality rate of 18.8%, was reported. No serious adverse events were reported. Finally, some practical and financial considerations are provided, which point to the therapy's recommendation as the first line of treatment for this particular and crucial patient group.
Collapse
Affiliation(s)
- Laura Rehak
- Athena Cell Therapy Technologies, 50126 Florence, Italy
| | - Laura Giurato
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Matteo Monami
- Department of Diabetology Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy
| | - Marco Meloni
- Diabetic Foot Unit, Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessia Scatena
- Diabetology Unit, San Donato Hospital Arezzo, Local Health Authorities Southeast Tuscany, 52100 Arezzo, Italy
| | - Andrea Panunzi
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
- PhD School of Applied Medical and Surgical Sciences, University of Rome Tor Vergata Italy, 00133 Rome, Italy
| | | | | | - Luigi Uccioli
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| |
Collapse
|
37
|
Weng Y, Feng Y, Li Z, Xu S, Wu D, Huang J, Wang H, Wang Z. Zfp260 choreographs the early stage osteo-lineage commitment of skeletal stem cells. Nat Commun 2024; 15:10186. [PMID: 39582024 PMCID: PMC11586402 DOI: 10.1038/s41467-024-54640-0] [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: 06/21/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
The initial fine-tuning processes are crucial for successful bone regeneration, as they guide skeletal stem cells through progenitor differentiation toward osteo- or chondrogenic fate. While fate determination processes are well-documented, the mechanisms preceding progenitor commitment remain poorly understood. Here, we identified a transcription factor, Zfp260, as pivotal for stem cell maturation into progenitors and directing osteogenic differentiation. Zfp260 is markedly up-regulated as cells transition from stem to progenitor stages; its dysfunction causes lineage arrest at the progenitor stage, impairing bone repair. Zfp260 is required for maintaining chromatin accessibility and regulates Runx2 expression by forming super-enhancer complexes. Furthermore, the PKCα kinase phosphorylates Zfp260 at residues Y173, S182, and S197, which are essential for its functional activity. Mutations at these residues significantly impair its functionality. These findings position Zfp260 as a vital factor bridging stem cell activation with progenitor cell fate determination, unveiling a element fundamental to successful bone regeneration.
Collapse
Affiliation(s)
- Yuteng Weng
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Yanhuizhi Feng
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Zeyuan Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Shuyu Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Di Wu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Jie Huang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Haicheng Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Zuolin Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China.
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
38
|
Tyrina E, Yakubets D, Markina E, Buravkova L. Hippo Signaling Pathway Involvement in Osteopotential Regulation of Murine Bone Marrow Cells Under Simulated Microgravity. Cells 2024; 13:1921. [PMID: 39594669 PMCID: PMC11592674 DOI: 10.3390/cells13221921] [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: 10/14/2024] [Revised: 11/02/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The development of osteopenia is one of the most noticeable manifestations of the adverse effects of space factors on crew members. The Hippo signaling pathway has been shown to play a central role in regulating the functional activity of cells through their response to mechanical stimuli. In the present study, the components of the Hippo pathway and the protective properties of osteodifferentiation inducers were investigated under simulated microgravity (smg) using a heterotypic bone marrow cell culture model, which allows for the maintenance of the close interaction between the stromal and hematopoietic compartments, present in vivo and of great importance for both the fate of osteoprogenitors and hematopoiesis. After 14 days of smg, the osteopotential and osteodifferentiation of bone marrow stromal progenitor cells, the expression of Hippo cascade genes and the immunocytochemical status of the adherent fraction of bone marrow cells, as well as the paracrine profile in the conditioned medium and the localization of Yap1 and Runx2 in mechanosensitive cells of the bone marrow were obtained. Simulated microgravity negatively affects stromal and hematopoietic cells when interacting in a heterotypic murine bone marrow cell culture. This is evidenced by the decrease in cell proliferation and osteopotential. Changes in the production of pleiotropic cytokines IL-6, GROβ and MCP-1 were revealed. Fourteen days of simulated microgravity induced a decrease in the nuclear translocation of Yap1 and the transcription factor Runx2 in the stromal cells of the intact group. Exposure to osteogenic induction conditions partially compensated for the negative effect of simulated microgravity. The data obtained will be crucial for understanding the effects of spaceflight on osteoprogenitor cell growth and differentiation via Hippo-Yap signaling.
Collapse
Affiliation(s)
- Ekaterina Tyrina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (D.Y.); (L.B.)
| | | | - Elena Markina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (D.Y.); (L.B.)
| | | |
Collapse
|
39
|
Shi Y, Gu J, Zhang C, Mi R, Ke Z, Xie M, Jin W, Shao C, He Y, Shi J, Xie Z. A Janus Microsphere Delivery System Orchestrates Immunomodulation and Osteoinduction by Fine-tuning Release Profiles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403835. [PMID: 38984921 DOI: 10.1002/smll.202403835] [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: 05/12/2024] [Revised: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Bone regeneration is a well-orchestrated process synergistically involving inflammation, angiogenesis, and osteogenesis. Therefore, an effective bone graft should be designed to target multiple molecular events and biological demands during the bone healing process. In this study, a biodegradable gelatin methacryloyl (GelMA)-based Janus microsphere delivery system containing calcium phosphate oligomer (CPO) and bone morphogenetic protein-2 (BMP-2) is developed based on natural biological events. The exceptional adjustability of GelMA facilitates the controlled release and on-demand application of biomolecules, and optimized delivery profiles of CPO and BMP-2 are explored. The sustained release of CPO during the initial healing stages contributes to early immunomodulation and promotes mineralization in the late stage. Meanwhile, the administration of BMP-2 at a relatively high concentration within the therapeutic range enhances the osteoinductive property. This delivery system, with fine-tuned release patterns, induces M2 macrophage polarization and creates a conducive immuno-microenvironment, which in turn facilitates effective bone regeneration in vivo. Collectively, this study proposes a bottom-up concept, aiming to develop a user-friendly and easily controlled delivery system targeting individual biological events, which may offer a new perspective on developing function-optimized biomaterials for clinical use.
Collapse
Affiliation(s)
- Yang Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jingyi Gu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Chun Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Rui Mi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhiwei Ke
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Mingjun Xie
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Wenjing Jin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Changyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- The Second Affiliated Hospital of Zhejiang University and State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310027, China
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
40
|
Cheng M, Liang X, Cui L, Guan D, Qu Y, Zhao J, Guan K. Magnesium-based nanocomposites for orthopedic applications: A review. JOURNAL OF MAGNESIUM AND ALLOYS 2024; 12:4335-4362. [DOI: 10.1016/j.jma.2024.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
|
41
|
Jiang K, Luo C, Li YM, Wang K, Huang S, You XH, Liu Y, Luo E, Xu JZ, Zhang L, Li ZM. An immunomodulatory and osteogenic bacterial cellulose scaffold for bone regeneration via regulating the immune microenvironment. Int J Biol Macromol 2024; 281:136375. [PMID: 39383912 DOI: 10.1016/j.ijbiomac.2024.136375] [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: 05/11/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Creating a bone homeostasis microenvironment that balances osteogenesis and immunity is a substantial challenge for bone regeneration. Here, we prepared an immunomodulatory and osteogenic bacterial cellulose scaffold (FOBS) via a facile one-pot approach. The aldehyde groups were generated via selective oxidation of the hydroxyl groups of bacterial cellulose, offering the bonding sites for dopamine through a Schiff base reaction. At the same time, the deposition of Ca2+ and PO43- was promoted on the aldehyde cellulose scaffold because of the high affinity of the catechol moiety for Ca2+. Compared with that of the unmodified scaffold, the hydroxyapatite content of FOBS increased by 47.1 % according to the ICP results. Interestingly, FOBS regulated the immune microenvironment to accelerate the conversion of M1 to M2 macrophages. The expressions of ARG-1 and Dectin-1 (M2) in the FOBS group increased by >100 %. The expression of osteogenic differentiation of BMSCs was also upregulated. In a rat cranial defect model, the BV/TV of FOBS was significantly increased. Further immunohistochemical analysis revealed that an improved immune microenvironment promoted the osteogenic differentiation of stem cells in vivo. This work provides an effective and easy-to-operate strategy for the development of the bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Kai Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chuan Luo
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yuan-Min Li
- Key Laboratory of Transplant Engineering and Immunology, NHC, Chengdu 610065, China; Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Kai Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shishu Huang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Xuan-He You
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Li Zhang
- Department of Rehabilitation Medicine, West China Second Hospital, Sichuan University, Chengdu 610065, China.
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
42
|
Zhao X, Wang Q, Wang W, Lu S. Increased neutrophil extracellular traps caused by diet-induced obesity delay fracture healing. FASEB J 2024; 38:e70126. [PMID: 39446097 PMCID: PMC11580727 DOI: 10.1096/fj.202401523r] [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: 07/03/2024] [Revised: 09/28/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Obesity, recognized as a risk factor for nonunion, detrimentally impacts bone health, with significant physical and economic repercussions for affected individuals. Nevertheless, the precise pathomechanisms by which obesity impairs fracture healing remain insufficiently understood. Multiple studies have identified neutrophil granulocytes as key players in the systemic immune response, being the predominant immune cells in early fracture hematomas. This study identified a previously unreported critical period for neutrophil infiltration into the callus. In vivo experiments demonstrated that diet-induced obesity (DIO) mice showed earlier neutrophil infiltration, along with increased formation of neutrophil extracellular traps (NETs), compared to control mice during the endochondral phase of fracture repair. Furthermore, Padi4 knockout was found to reduce NET formation and mitigate the fracture healing delays caused by high-fat diets. Mechanistically, in vitro analyses revealed that NETs, by activating NLRP3 inflammasomes, inhibited the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and concurrently promoted M1-like macrophage polarization. These findings establish a connection between NET formation during the endochondral phase and delayed fracture healing, suggesting that targeting NETs could serve as a promising therapeutic approach for addressing obesity-induced delays in fracture recovery.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Orthopedics, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
| | - Qijun Wang
- Department of Orthopedics, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
| | - Wei Wang
- Department of Orthopedics, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
| | - Shibao Lu
- Department of Orthopedics, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
| |
Collapse
|
43
|
Wang X, Wang S, Mu H, Yang C, Dong W, Wang X, Wang J. Macrophage-derived amphiregulin promoted the osteogenic differentiation of chondrocytes through EGFR/Yap axis and TGF-β activation. Bone 2024; 190:117275. [PMID: 39383984 DOI: 10.1016/j.bone.2024.117275] [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: 02/25/2024] [Revised: 07/15/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Endochondral ossification represents a crucial biological process in skeletal development and bone defect repair. Macrophages, recognized as key players in the immune system, are now acknowledged for their substantial role in promoting endochondral ossification within cartilage. Concurrently, the epidermal growth factor receptor (EGFR) ligand amphiregulin (Areg) has been documented for its contributory role in restoring bone tissue homeostasis post-injury. However, the mechanism by which macrophage-secreted Areg facilitates bone repair remains elusive. In this study, the induction of macrophage depletion through in vivo administration of clodronate liposomes was employed in a standard open tibial fracture mouse model to assess bone healing using micro-computed tomography (micro-CT) analysis, histomorphology, and ELISA serum evaluations. The investigation revealed sustained expression of Areg during the fracture healing period in wild-type mice. Macrophage depletion significantly reduced the number of macrophages on the local bone surface and vital organs. This reduction led to diminished Areg secretion, decreased collagen production, and delayed fracture healing. However, histological and micro-CT assessments at 7 and 21 days post-local Areg treatment exhibited a marked improvement of bone healing compared to the vehicle control. In vitro studies demonstrated an increase of Areg secretion by the Raw264.7 cells upon ATP stimulation. Indirect co-culture of Raw264.7 and ATDC5 cells indicated that Areg overexpression enhanced the osteogenic potential of chondrocytes, and vice versa. This osteogenic promotion was attributed to Areg's activation of the membrane receptor EGFR in the ATDC5 cell line, the enhanced phosphorylation of transcription factor Yap, and the facilitation of the expression of bioactive TGF-β by chondrocytes. Collectively, this research elucidates the direct mechanistic effects of macrophage-secreted Areg in promoting bone homeostasis following bone injury.
Collapse
Affiliation(s)
- Xinyi 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, Hubei 430079, China
| | - Shuo 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, Hubei 430079, China
| | - Hailin Mu
- 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, Hubei 430079, China
| | - Chang Yang
- 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, Hubei 430079, China
| | - Wei Dong
- 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, Hubei 430079, China
| | - Xinru 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, Hubei 430079, China
| | - Jaiwei 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, Hubei 430079, China.
| |
Collapse
|
44
|
Batoon L, Keshvari S, Irvine KM, Ho E, Caruso M, Patkar OL, Sehgal A, Millard SM, Hume DA, Pettit AR. Relative contributions of osteal macrophages and osteoclasts to postnatal bone development in CSF1R-deficient rats and phenotype rescue following wild-type bone marrow cell transfer. J Leukoc Biol 2024; 116:753-765. [PMID: 38526212 DOI: 10.1093/jleuko/qiae077] [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: 09/29/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Macrophage and osteoclast proliferation, differentiation and survival are regulated by colony-stimulating factor 1 receptor (CSF1R) signaling. Osteopetrosis associated with Csf1 and Csf1r mutations has been attributed to the loss of osteoclasts and deficiency in bone resorption. Here, we demonstrate that homozygous Csf1r mutation in rat leads to delayed postnatal skeletal ossification associated with substantial loss of osteal macrophages in addition to osteoclasts. Osteosclerosis and site-specific skeletal abnormalities were reversed by intraperitoneal transfer of wild-type bone marrow cells (bone marrow cell transfer, BMT) at weaning. Following BMT, IBA1+ macrophages were detected before TRAP+ osteoclasts at sites of ossification restoration. These observations extend evidence that osteal macrophages independently contribute to bone anabolism and are required for normal postnatal bone growth and morphogenesis.
Collapse
Affiliation(s)
- Lena Batoon
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Sahar Keshvari
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Katharine M Irvine
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Eileen Ho
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Melanie Caruso
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Omkar L Patkar
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Anuj Sehgal
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Susan M Millard
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - David A Hume
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Allison R Pettit
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| |
Collapse
|
45
|
Daneshvar A, Nemati P, Azadi A, Amid R, Kadkhodazadeh M. M2 macrophage-derived exosomes for bone regeneration: A systematic review. Arch Oral Biol 2024; 166:106034. [PMID: 38943857 DOI: 10.1016/j.archoralbio.2024.106034] [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: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVE This systematic review aims to evaluate existing evidence to investigate the therapeutic efficacy of M2 macrophage-derived exosomes in bone regeneration. DESIGN A comprehensive search between 2020 and 2024 across PubMed, Web of Science, and Scopus was conducted using a defined search strategy to identify relevant studies regarding the following question: "What is the impact of M2 macrophage-derived exosomes on bone regeneration?". Controlled in vitro and in vivo studies were included in this study. The SYRCLE tool was used to evaluate the risk of bias in the included animal studies. RESULTS This review included 20 studies published. Seven studies were selected for only in vitro analysis, whereas 13 studies underwent both in vitro and in vivo analyses. The in vivo studies employed animal models, including 163 C57BL6 mice and 73 Sprague-Dawley rats. Exosomes derived from M2 macrophages were discovered to be efficacious in promoting bone regeneration and vascularization in animal models of bone defects. These effects were primarily confirmed through morphological and histological assessments. This remarkable outcome is attributed to the regulation of multiple signaling pathways, as evidenced by the findings of 11 studies investigating the involvement of miRNAs in this intricate process. In addition, in vitro studies observed positive effects on cell proliferation, migration, osteogenesis, and angiogenesis. Heterogeneity in study methods hinders direct comparison of results across studies. CONCLUSION M2 macrophage-derived exosomes demonstrate remarkable potential for promoting bone regeneration. Further research optimizing their application and elucidating the underlying mechanisms can pave the way for clinical translation.
Collapse
Affiliation(s)
- Alireza Daneshvar
- Student Research Committee, Faculty of Dentistry, Islamic Azad University, Tehran, Iran
| | - Parisa Nemati
- Student Research Committee, Faculty of Dentistry, Islamic Azad University, Tehran, Iran
| | - Ali Azadi
- Dentofacial Deformities Research Center, Research Institute for Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Amid
- Department of Periodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Kadkhodazadeh
- Department of Periodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Dental Research Center, Research Institute for Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Chow SKH, Gao Q, Pius A, Morita M, Ergul Y, Murayama M, Shinohara I, Cekuc MS, Ma C, Susuki Y, Goodman SB. The Advantages and Shortcomings of Stem Cell Therapy for Enhanced Bone Healing. Tissue Eng Part C Methods 2024; 30:415-430. [PMID: 39311464 DOI: 10.1089/ten.tec.2024.0252] [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] [Indexed: 10/05/2024] Open
Abstract
This review explores the regenerative potential of key progenitor cell types and therapeutic strategies to improve healing of complex fractures and bone defects. We define, summarize, and discuss the differentiation potential of totipotent, pluripotent, and multipotent stem cells, emphasizing the advantages and shortcomings of cell therapy for bone repair and regeneration. The fundamental role of mesenchymal stem cells is highlighted due to their multipotency to differentiate into the key lineage cells including osteoblasts, osteocytes, and chondrocytes, which are crucial for bone formation and remodeling. Hematopoietic stem cells (HSCs) also play a significant role; immune cells such as macrophages and T-cells modulate inflammation and tissue repair. Osteoclasts are multinucleated cells that are important to bone remodeling. Vascular progenitor (VP) cells are critical to oxygen and nutrient supply. The dynamic interplay among these lineages and their microenvironment is essential for effective bone restoration. Therapies involving cells that are more than "minimally manipulated" are controversial and include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). ESCs, derived from early-stage embryos, possess pluripotent capabilities and have shown promise in preclinical studies for bone healing. iPSCs, reprogrammed from somatic cells, offer personalized medicine applications and can differentiate into various tissue-specific cell lines. Minimally manipulative cell therapy approaches such as the use of bone marrow aspirate concentrate (BMAC), exosomes, and various biomaterials for local delivery are explored for their effectiveness in bone regeneration. BMAC, which contains mostly immune cells but few mesenchymal and VPs, probably improves bone healing by facilitating paracrine-mediated intercellular communication. Exosome isolation harnesses the biological signals and cellular by-products that are a primary source for cell crosstalk and activation. Safe, efficacious, and cost-effective strategies to enhance bone healing using novel cellular therapies are part of a changing paradigm to modulate the inflammatory, repair, and regenerative pathways to achieve earlier more robust tissue healing and improved physical function. Impact Statement Stem cell therapy holds immense potential for bone healing due to its ability to regenerate damaged tissue. Nonmanipulated bone marrow aspirate contains mesenchymal stem cells that promote bone repair and reduce healing time. Induced pluripotent stem cells offer the advantage of creating patient-specific cells that can differentiate into osteoblasts, aiding in bone regeneration. Other delivery methods, such as scaffold-based techniques, enhance stem cell integration and function. Collectively, these approaches can improve treatment outcomes, reduce recovery periods, and advance our understanding of bone healing mechanisms, making them pivotal in orthopedic research and regenerative medicine.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Alexa Pius
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yasemin Ergul
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mehmet Sertac Cekuc
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chao Ma
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
47
|
Hatano E, Akhter N, Anada R, Ono M, Oohashi T, Kuboki T, Kamioka H, Okada M, Matsumoto T, Hara ES. The cell membrane as biofunctional material for accelerated bone repair. Acta Biomater 2024; 186:411-423. [PMID: 39089349 DOI: 10.1016/j.actbio.2024.07.037] [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: 03/03/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/03/2024]
Abstract
The cell (plasma) membrane is enriched with numerous receptors, ligands, enzymes, and phospholipids that play important roles in cell-cell and cell-extracellular matrix interactions governing, for instance, tissue development and repair. We previously showed that plasma membrane nanofragments (PMNFs) act as nucleation sites for bone formation in vivo, and induce in vitro mineralization within 1 day. In this study, we optimized the methods for generating, isolating, and applying PMNFs as a cell-free therapeutic to expedite bone defect repair. The PMNFs were isolated from different mouse cell lines (chondrocytes, osteoblasts, and fibroblasts), pre-conditioned, lyophilized, and subsequently transplanted into 2 mm critical-sized calvarial defects in mice (n = 75). The PMNFs from chondrocytes, following a 3-day pre-incubation, significantly accelerated bone repair within 2 weeks, through a coordinated attraction of macrophages, endothelial cells, and osteoblasts to the healing site. In vitro experiments confirmed that PMNFs enhanced cell adhesion. Comparison of the PMNF efficacy with phosphatidylserine, amorphous calcium phosphate (ACP), and living cells confirmed the unique ability of PMNFs to promote accelerated bone repair. Importantly, PMNFs promoted nearly complete integration of the regenerated bone with native tissue after 6 weeks (% non-integrated bone area = 15.02), contrasting with the partial integration (% non-integrated bone area = 56.10; p < 0.01, Student's test) with transplantation of ACP. Vickers microhardness tests demonstrated that the regenerated bone after 6 weeks (30.10 ± 1.75) exhibited hardness similar to native bone (31.07 ± 2.46). In conclusion, this is the first study to demonstrate that cell membrane can be a promising cell-free material with multifaceted biofunctional properties that promote accelerated bone repair. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Emi Hatano
- Advanced Research Center for Oral and Craniofacial Sciences Dental School, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nahid Akhter
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Risa Anada
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Okada
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Emilio Satoshi Hara
- Advanced Research Center for Oral and Craniofacial Sciences Dental School, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
48
|
Ge R, Liu C, Zhao Y, Wang K, Wang X. Endochondral Ossification for Spinal Fusion: A Novel Perspective from Biological Mechanisms to Clinical Applications. J Pers Med 2024; 14:957. [PMID: 39338212 PMCID: PMC11433020 DOI: 10.3390/jpm14090957] [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: 06/04/2024] [Revised: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Degenerative scoliosis (DS), encompassing conditions like spondylolisthesis and spinal stenosis, is a common type of spinal deformity. Lumbar interbody fusion (LIF) stands as a conventional surgical intervention for this ailment, aiming at decompression, restoration of intervertebral height, and stabilization of motion segments. Despite its widespread use, the precise mechanism underlying spinal fusion remains elusive. In this review, our focus lies on endochondral ossification for spinal fusion, a process involving vertebral development and bone healing. Endochondral ossification is the key step for the successful vertebral fusion. Endochondral ossification can persist in hypoxic conditions and promote the parallel development of angiogenesis and osteogenesis, which corresponds to the fusion process of new bone formation in the hypoxic region between the vertebrae. The ideal material for interbody fusion cages should have the following characteristics: (1) Good biocompatibility; (2) Stable chemical properties; (3) Biomechanical properties similar to bone tissue; (4) Promotion of bone fusion; (5) Favorable for imaging observation; (6) Biodegradability. Utilizing cartilage-derived bone-like constructs holds promise in promoting bony fusion post-operation, thus warranting exploration in the context of spinal fusion procedures.
Collapse
Affiliation(s)
- Rile Ge
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Rd, Beijing 100050, China;
| | - Chenjun Liu
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Yuhong Zhao
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| | - Kaifeng Wang
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Xiluan Wang
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| |
Collapse
|
49
|
Díez-Tercero L, Bosch-Rué È, Bosch BM, Rojas-Márquez R, Caballé-Serrano J, Delgado LM, Pérez RA. Engineering a microparticle-loaded rough membrane for guided bone regeneration modulating osteoblast response without inducing inflammation. Colloids Surf B Biointerfaces 2024; 241:113994. [PMID: 38850744 DOI: 10.1016/j.colsurfb.2024.113994] [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: 03/12/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Guided bone regeneration (GBR) is a widely used procedure that prevents the fast in-growth of soft tissues into bone defect. Among the different types of membranes, the use of collagen membranes is the gold standard. However, these membranes are implanted in tissue location where a severe acute inflammation will occur and can be negatively affected. The aim of this study was to develop a collagen-based membrane for GBR that incorporated alginate-hydroxyapatite microparticles. Membranes were manufactured using collagen type I and gelatin and alginate-hydroxyapatite microparticles. Membranes were assessed in terms of topography by scanning electron microscopy and confocal microscopy; stability by swelling after an overnight incubation in saline and enzymatic degradation against collagenase and mechanical properties by tensile tests. Furthermore, the biological response was assessed with SaOs-2 cells and THP-1 macrophages to determine alkaline phosphatase activity and inflammatory cytokine release. Our results showed that the incorporation of different percentages of these microparticles could induce changes in the surface topography. When the biological response was analyzed, either membranes were not cytotoxic to THP-1 macrophages or to SaOs-2 cells and they did not induce the release of pro-inflammatory cytokines. However, the different surface topographies did not induce changes in the macrophage morphology and the release of pro- and anti-inflammatory cytokines, suggesting that the effect of surface roughness on macrophage behavior could be dependent on other factors such as substrate stiffness and composition. Collagen-gelatin membranes with embedded alginate-hydroxyapatite microparticles increased ALP activity, suggesting a positive effect of them on bone regeneration, remaining unaffected the release of pro- and anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Èlia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Begoña M Bosch
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Raquel Rojas-Márquez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Jordi Caballé-Serrano
- Department of Oral and Maxillofacial Surgery, Universitat Internacional de Catalunya, Barcelona, Spain; Department of Periodontology, School of Dental Medicine - University of Bern, Bern, Switzerland
| | - Luis M Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| | - Román A Pérez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| |
Collapse
|
50
|
Yu T, Wang J, Zhou Y, Ma C, Bai R, Huang C, Wang S, Liu K, Han B. Harnessing Engineered Extracellular Vesicles from Mesenchymal Stem Cells as Therapeutic Scaffolds for Bone‐Related Diseases. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202402861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Indexed: 10/05/2024]
Abstract
AbstractMesenchymal stem cells (MSCs) play a crucial role in maintaining bone homeostasis and are extensively explored for cell therapy in various bone‐related diseases. In addition to direct cell therapy, the secretion of extracellular vesicles (EVs) by MSCs has emerged as a promising alternative approach. MSC‐derived EVs (MSC‐EVs) offer equivalent therapeutic efficacy to MSCs while mitigating potential risks. These EVs possess unique properties that enable them to traverse biological barriers and deliver bioactive cargos to target cells. Furthermore, by employing modification and engineering strategies, the therapeutic effects and tissue targeting specificity of MSC‐EVs can be further enhanced to meet specific therapeutic needs. In this review, the mechanisms and advantages of MSC‐EV therapy in diseased bone tissues are highlighted. Through simple isolation and modification techniques, MSC‐EV‐based biomaterials have demonstrated great promise for bone regeneration. Finally, future perspectives on MSC‐EV therapy are presented, envisioning the development of next‐generation regenerative materials and bioactive agents for clinical translation in the field of bone regeneration.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Jingwei Wang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Yusai Zhou
- School of Materials Science and Engineering Beihang University Beijing 100191 P. R. China
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Rushui Bai
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Cancan Huang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Shidong Wang
- Musculoskeletal Tumor Center Peking University People's Hospital No.11 Xizhimen South St. Beijing 100044 P. R. China
| | - Kai Liu
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Bing Han
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| |
Collapse
|