1
|
Yang L, Bai X, Liu Y, Zhu S, Li S, Chen Z, Han T, Jin S, Zang M. Angiosome-Guided Perfusion Decellularization of Fasciocutaneous Flaps. J Reconstr Microsurg 2025; 41:405-414. [PMID: 39191422 DOI: 10.1055/a-2404-2608] [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: 08/29/2024]
Abstract
BACKGROUND Tissue engineering based on whole-organ perfusion decellularization has successfully generated small-animal organs, including the heart and limbs. Herein, we aimed to use angiosome-guided perfusion decellularization to develop an acellular fasciocutaneous flap matrix with an intact vascular network. METHODS Abdominal flaps of rats were harvested, and the vascular pedicle (iliac artery and vein) was dissected and injected with methylene blue to identify the angiosome region and determine the flap dimension for harvesting. To decellularize flaps, the iliac artery was perfused sequentially with 1% sodium dodecyl sulfate (SDS), deionized water, and 1% Triton-X100. Gross morphology, histology, and DNA quantity of flaps were then obtained. Flaps were also subjected to glycosaminoglycan (GAG) and hydroxyproline content assays and computed tomography angiography. RESULTS Histological assessment indicated that cellular content was completely removed in all flap layers following a 10-hour perfusion in SDS. DNA quantification confirmed 81% DNA removal. Based on biochemical assays, decellularized flaps had hydroxyproline content comparable with that of native flaps, although significantly fewer GAGs (p = 0.0019). Histology and computed tomography angiography illustrated the integrity and perfusability of the vascular system. CONCLUSION The proposed angiosome-guided perfusion decellularization protocol could effectively remove cellular content from rat fasciocutaneous flaps and preserve the integrity of innate vascular networks.
Collapse
Affiliation(s)
- Liya Yang
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xueshan Bai
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yuanbo Liu
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shan Zhu
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shanshan Li
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zixiang Chen
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Tinglu Han
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shengyang Jin
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Mengqing Zang
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
2
|
Liu X, Hu H, Ma J, Wang B. Mineralized cellulose nanofibers reinforced bioactive hydrogel remodels the osteogenic and angiogenic microenvironment for enhancing bone regeneration. Carbohydr Polym 2025; 357:123480. [PMID: 40159001 DOI: 10.1016/j.carbpol.2025.123480] [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: 01/03/2025] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025]
Abstract
Slow osteogenesis and insufficient vascularization remain significant challenges in achieving effective bone repair and functional restoration with tissue-engineered scaffolds. Herein, a novel mineralized nanofibers reinforced bioactive hydrogel was designed to enhance bone regeneration inspired from the structural and functional properties of the bone tissue extracellular matrix (ECM). This bioactive hydrogel integrated enzymatically mineralized TEMPO-oxidized bacterial cellulose (m-TOBC) nanofibers and mesoporous silica nanoparticles (MSNs) loaded with the angiogenic drug dimethyloxalylglycine (DMOG) into gelatin methacryloyl (GelMA). The m-TOBC nanofibers achieved one stone, three birds: improving the printability of GelMA ink, mechanical properties, and osteoconduction of the hydrogel. The incorporation of MSNs loaded with DMOG fostered an angiogenic microenvironment through the release of DMOG. Results indicated that the bioactive hydrogel significantly enhanced in vitro mineralized matrix deposition and osteoblastic alkaline phosphatase expression. Additionally, the bioactive hydrogel had good ability to promote angiogenesis in terms of enhanced endothelial cell migration, tube formation, and upregulated angiogenic genes expression levels. In a critical-sized rat cranial defect model, the bioactive hydrogel significantly enhanced bone regeneration. Overall, this research offered a promising strategy to design nanofibers enhanced hydrogel to remodel osteogenic and angiogenic microenvironment for enhancing bone repair.
Collapse
Affiliation(s)
- Xiaokang Liu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Haoran Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jinghong Ma
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Baoxiu Wang
- School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
3
|
Hu Y, Li M, Xu X, Ma N, Luo J, Wu X, Ping Q, Lin X, Zhang T, Liang C, Yang L. A bioactive Cu-grafted gel coating with micro-nano structures for simultaneous enhancement of bone regeneration and infection resistance. J Mater Chem B 2025. [PMID: 40391950 DOI: 10.1039/d5tb00211g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Prosthetic joint infection (PJI) remains a significant challenge in clinical applications. It not only impedes the recovery of bone tissue at the site of bone defect but also leads to multiple debridements, long-lasting antibiotic treatment and even secondary replacement. Titanium alloy Ti6Al4V (TC4) is widely used in orthopedic implants due to its excellent mechanical properties and biocompatibility; however, it lacks inherent antibacterial and osteoinductive functions. In this study, a composite coating based on polyvinyl alcohol (PVA) with tissue repair and antibacterial properties was applied on the surface of TC4. A PVA gel coating functionalized with terpyridine and catechol groups (PVA-TP-CA) was synthesized and subsequently complexed with copper (Cu) ions. The differential binding affinities of TP and CA groups to Cu enabled a sustained and controlled release of metal ions. Furthermore, a micro-nano surface structure was fabricated on TC4 using femtosecond laser technology to achieve a micro-nano structure interface and enhanced bonding strength. Biological evaluations demonstrated that the modified surface significantly improved the antibacterial, angiogenic, and osteogenic properties of TC4. These findings indicate that this multifunctional composite coating holds great promise for surface modification of orthopedic implants, offering an effective strategy for preventing PJI while promoting bone regeneration.
Collapse
Affiliation(s)
- Ying Hu
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Mingjun Li
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Kantstr. 55, 14513 Teltow, Germany
| | - Nan Ma
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Jiahao Luo
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Xiaoxuan Wu
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Qixiang Ping
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Xiao Lin
- Orthopedic Institute, Department of Orthopedics, the First Affiliated Hospital, Soochow University, Suzhou, China
| | - Tingbin Zhang
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Chunyong Liang
- Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300130, China.
| | - Lei Yang
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China.
| |
Collapse
|
4
|
Jiang J, Wang J, Fan P, Zhao Z, Deng H, Li J, Wang Y, Wang Y. Biomaterial-based strategies for bone cement: modulating the bone microenvironment and promoting regeneration. J Nanobiotechnology 2025; 23:343. [PMID: 40361125 PMCID: PMC12070552 DOI: 10.1186/s12951-025-03363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoporotic bone defect and fracture healing remain significant challenges in clinical practice. While traditional therapeutic approaches provide some regulation of bone homeostasis, they often present limitations and adverse effects. In orthopedic procedures, bone cement serves as a crucial material for stabilizing osteoporotic bone and securing implants. However, with the exception of magnesium phosphate cement, most cement variants lack substantial bone regenerative properties. Recent developments in biomaterial science have opened new avenues for enhancing bone cement functionality through innovative modifications. These advanced materials demonstrate promising capabilities in modulating the bone microenvironment through their distinct physicochemical properties. This review provides a systematic analysis of contemporary biomaterial-based modifications of bone cement, focusing on their influence on the bone healing microenvironment. The discussion begins with an examination of bone microenvironment pathology, followed by an evaluation of various biomaterial modifications and their effects on cement properties. The review then explores regulatory strategies targeting specific microenvironmental elements, including inflammatory response, oxidative stress, osteoblast-osteoclast homeostasis, vascular network formation, and osteocyte-mediated processes. The concluding section addresses current technical challenges and emerging research directions, providing insights for the development of next-generation biomaterials with enhanced functionality and therapeutic potential.
Collapse
Affiliation(s)
- Jiawei Jiang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Juan Wang
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, Jiangsu, China
| | - Pan Fan
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhe Zhao
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China
| | - Hongjian Deng
- Department of Orthopaedics, The Affiliated 2 Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian Li
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| | - Yi Wang
- Department of Orthopaedics, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| | - Yuntao Wang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| |
Collapse
|
5
|
Wang B, Lyu FJ, Deng Z, Zheng Q, Ma Y, Peng Y, Guo S, Lei G, Lai Y, Li Q. Therapeutic potential of stem cell-derived exosomes for bone tissue regeneration around prostheses. J Orthop Translat 2025; 52:85-96. [PMID: 40291635 PMCID: PMC12023751 DOI: 10.1016/j.jot.2025.03.007] [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: 10/10/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Artificial joint replacement is a widely recognized treatment for arthritis and other severe joint conditions. However, one of the primary causes of failure in joint replacements is the loosening of the prosthesis. After implantation, wear particles between the implant and the adjacent bone tissue are the principal contributors to this loosening. Recently, exosomes have garnered significant interest due to their low immunogenicity and effective membrane binding. They have shown potential in promoting bone regeneration via the paracrine pathway. This review examines the role and mechanisms of exosomes derived from mesenchymal stem cells (MSCs) in bone regeneration, their impact on the integration of various implants into surrounding bone tissue and current challenges and future directions for the clinical application of exosomes. The Translational Potential of this Article: Emerging evidence suggests that mesenchymal stem cell-derived exosomes may offer a promising therapeutic strategy for aseptic prosthesis loosening, potentially mediated through mechanisms such as modulation of inflammatory responses, suppression of osteoclastogenesis, enhancement of osteogenic differentiation and facilitation of bone regeneration. Preclinical studies further indicate that the therapeutic potential of these extracellular vesicles could be optimized through bioengineering strategies, including surface modification and cargo-loading techniques, warranting further investigation to advance their clinical translation.
Collapse
Affiliation(s)
- Biwu Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yujie Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Shantou University Medical College, Xinling Road 22, Shantou, 515041, China
| | - Shujun Guo
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Guihua Lei
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yonggang Lai
- South China University of Technology-The University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qingtian Li
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
6
|
Sun J, Chen C, Zhang B, Yao C, Zhang Y. Advances in 3D-printed scaffold technologies for bone defect repair: materials, biomechanics, and clinical prospects. Biomed Eng Online 2025; 24:51. [PMID: 40301861 PMCID: PMC12042599 DOI: 10.1186/s12938-025-01381-w] [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: 12/25/2024] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
The treatment of large bone defects remains a significant clinical challenge due to the limitations of current grafting techniques, including donor site morbidity, restricted availability, and suboptimal integration. Recent advances in 3D bioprinting technology have enabled the fabrication of structurally and functionally optimized scaffolds that closely mimic native bone tissue architecture. This review comprehensively examines the latest developments in 3D-printed scaffolds for bone regeneration, focusing on three critical aspects: (1) material selection and composite design encompassing metallic; (2) structural optimization with hierarchical porosity (macro/micro/nano-scale) and biomechanical properties tailored; (3) biological functionalization through growth factor delivery, cell seeding strategies and surface modifications. We critically analyze scaffold performance metrics from different research applications, while discussing current translational barriers, including vascular network establishment, mechanical stability under load-bearing conditions, and manufacturing scalability. The review concludes with a forward-looking perspective on innovative approaches such as 4D dynamic scaffolds, smart biomaterials with stimuli-responsive properties, and the integration of artificial intelligence for patient-specific design optimization. These technological advancements collectively offer unprecedented opportunities to address unmet clinical needs in complex bone reconstruction.
Collapse
Affiliation(s)
- Jie Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Cao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Bo Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Chen Yao
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Yafeng Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
7
|
Artamonov MY, Sokov EL, Kornilova LE, Minenko IA. The Intraosseous Environment: Physiological Parameters, Regulatory Mechanisms, and Emerging Insights in Bone Biology. Int J Mol Sci 2025; 26:3876. [PMID: 40332558 PMCID: PMC12027868 DOI: 10.3390/ijms26083876] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
The intraosseous environment is a dynamic and intricate system integral to bone health, encompassing vascular, cellular, and biochemical interactions that drive key processes such as hematopoiesis, bone remodeling, and systemic mineral regulation. This review examines the structural composition of the bone matrix, the diverse cellular landscape, and the interconnected vascular and nervous networks, highlighting their roles in preserving bone function and responding to pathological changes. Recent studies reveal regulatory mechanisms involving oxygen tension, ionic balance, signaling molecules, and mechanotransduction pathways that shape bone metabolism and its adaptation to mechanical forces. Insights into the bone microenvironment's metabolic shifts in cancer and its interaction with inflammation underscore its pivotal role in disease progression and therapeutic innovation. Additionally, advances in imaging techniques and biomaterials fuel progress in bone regeneration and understanding its microenvironment. Exploring the intricate physiochemical dynamics and regulatory networks within the intraosseous system unlocks potential clinical applications in bone diseases, tissue engineering, and systemic metabolic disorders. This comprehensive review bridges fundamental science with translational research, offering insights into the complex yet essential intraosseous environment.
Collapse
Affiliation(s)
- Mikhail Yu. Artamonov
- Department of Physical Medicine and Rehabilitation, Penn Medicine Princeton Health, Plainsboro, NJ 08536, USA
| | - Evgeniy L. Sokov
- Department of Algology, Peoples’ Friendship University, Moscow 117198, Russia
| | | | - Inessa A. Minenko
- Department of Sports Medicine, Sechenov Medical University, Moscow 119991, Russia;
| |
Collapse
|
8
|
Knothe Tate ML. The Flow of Life: Convergent Approaches to Understanding Musculoskeletal Health from Molecular- to Meso-Length Scales. FRONT BIOSCI-LANDMRK 2025; 30:25231. [PMID: 40302317 DOI: 10.31083/fbl25231] [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: 12/28/2023] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 05/02/2025]
Abstract
In the current perspective and review article, we address the human body as a living ecosystem with collecting watersheds and draining hydrosheds; we integrate our discoveries over the past quarter of a century and pose the critical open research questions to be addressed going forward, with the aim to improve cell, tissue, organ and organismal health. First, we address the flow of fluid through the tissues of the musculoskeletal system, after which we describe the interactions of the fluid, at multiple lengths and time scales, with the molecular to macroscopic non-fluid tissue components, discussing bone and tissues in the context of "living" chromatography and/or electrophoresis columns. Thereafter, we discuss the implications of functional barrier integrity, and the effects of cytokines on active barrier function and molecular transport between organ systems, tissue compartments, and within tissues. In addition, we address the fluid and its flow and the multi-physics implications thereof for the living inhabitants of tissues, i.e., the cells. Finally, we describe the implications of the solid and fluid components and the cellular inhabitants on ecosystem health, where the tissues and organs comprise the organism form interacting ecosystems throughout life and in the context of health and disease. By taking convergent approaches to understanding musculoskeletal, human and environmental health (which themselves are interdependent), we hope to pave new paths of innovation and discovery, to improve the lives of our worlds' inhabitants, from the worlds of our bone and joints and bodies to the interacting ecosystems of our Earth to unknown worlds beyond our current understanding.
Collapse
Affiliation(s)
- Melissa Louise Knothe Tate
- Blue Mountains World Interdisciplinary Innovation Institute, Blue Mountains National Park, NSW 2782, Australia
| |
Collapse
|
9
|
Zambuzzi WF, Ferreira MR. Dynamic ion-releasing biomaterials actively shape the microenvironment to enhance healing. J Trace Elem Med Biol 2025; 89:127657. [PMID: 40250222 DOI: 10.1016/j.jtemb.2025.127657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Dynamic ion-releasing biomaterials have redefined the role of implantable bone devices, transitioning them from passive mechanical support to active players in tissue regeneration. These materials actively modulate the surrounding biological microenvironment by releasing bioactive ions (e.g.: calcium, phosphate, and cobalt) which dynamically interact with cells and tissues surrounding them. This interaction becomes the microenvironment highly active and accelerates bone healing, promoting osteogenesis, and enhancing osseointegration. The ions modulate key biological processes in this regard, including osteoblast adhesion, proliferation, differentiation, angiogenesis, and immune responses, as well as coupled physiological mechanisms, ensuring that the implanted biomaterials foster an optimal environment for bone regeneration. More advanced surface modifications onto materials (e.g.: nanostructuring hydroxyapatites coatings) have been shown to further boost ion release, amplifying the ability of the material to influence surrounding tissues. As a result, ion-releasing biomaterials not only improve implant integration but also accelerate the overall healing process. Looking forward, the development of smart biomaterials capable of adjusting ion release in response to environmental changes offers exciting possibilities for personalized regenerative therapies and this review provides a comprehensive understanding of how dynamic ion-releasing biomaterials actively shape the microenvironment to enhance healing, focusing on their ability to modulate biological processes such as osteogenesis and angiogenesis. By examining the latest advances in surface modifications and ion-release mechanisms, this review also aims to revise the potential of these materials to revolutionize regenerative medicine, offering knowledge to guide the development of next-generation biomaterials for improved clinical outcomes.
Collapse
Affiliation(s)
- Willian Fernando Zambuzzi
- UNESP: São Paulo State University - Laboratory of Bioassays and Cellular Dynamics, Department of Chemical and Biological Science, Institute of Biosciences, Botucatu, São Paulo 18618-970, Brazil.
| | - Marcel Rodrigues Ferreira
- UNESP: São Paulo State University - Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit - Unipex, School of Medicine, Botucatu, São Paulo, Brazil
| |
Collapse
|
10
|
Fu FS, Chen HH, Chen Y, Yuan Y, Zhao Y, Yu A, Zhang XZ. Engineered bacillus subtilis enhances bone regeneration via immunoregulation and anti-Infection. Bioact Mater 2025; 46:503-515. [PMID: 39868074 PMCID: PMC11760808 DOI: 10.1016/j.bioactmat.2025.01.003] [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: 10/26/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Chronic osteomyelitis caused by implant infections is a common complication following orthopedic surgery. Preventing bacterial infection and simultaneously improving bone regeneration are the key for osteomyelitis. Current treatments include systemic antibiotics and multiple surgical interventions, but the strategies available for treatment are limited. In this study, a multifunctional engineered Bacillus subtilis (B. sub) hydrogel with sulfasalazine (SSZ) is developed to treat methicillin-resistant Staphylococcus aureus (MRSA) infection and anti-inflammatory and promote bone regeneration. B. sub in alginate hydrogels protects B. sub from being cleared by the host immune system while allowing the release of its bioactive substances, including antibacterial peptides and anti-inflammatory agents such as SSZ. The results show that the engineered probiotic hydrogels exhibit excellent antibacterial efficacy against MRSA (97 %) and prevent the development of bacterial resistance. The antibacterial effect is primarily mediated through the secretion of bioactive peptides by B. sub, which not only inhibit MRSA growth but also reduce the likelihood of resistance development. Meanwhile, the probiotic hydrogel has a greater ability to induce M2 polarization of macrophages and promote angiogenesis, resulting in enhanced osteogenic differentiation in bone marrow mesenchymal stem cells (BMSCs) and thus enhancing bone regeneration. This engineered probiotic hydrogel offers a promising strategy by simultaneously combating bacterial infection and enhancing osteogenic differentiation for chronic osteomyelitis.
Collapse
Affiliation(s)
- Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Huan-Huan Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, PR China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Ying Yuan
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Yong Zhao
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, PR China
| |
Collapse
|
11
|
Brandt Z, Nguyen K, Harianja G, Amin K, Battikha A, Betdashtoo N, Kubba R, Shin D, Oliinik M, Razzouk J, Chung JH, Danisa O, Cheng W. Location of Vascular Structures at Risk in Relation to Sacroiliac Joint Fusion. Spine (Phila Pa 1976) 2025; 50:493-499. [PMID: 40053916 DOI: 10.1097/brs.0000000000005218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/10/2024] [Indexed: 03/09/2025]
Abstract
STUDY DESIGN Retrospective cohort. OBJECTIVE This study seeks to establish the normal distribution of the vasculature surrounding the SI joint while also demonstrating associations between distribution and laterality, sex, and ethnicity. SUMMARY OF BACKGROUND DATA Sacroiliac (SI) joint fusion surgery has emerged as a viable treatment option for patients suffering from low back pain due to chronic SI joint dysfunction. Due to potential complications from iatrogenic injury to vasculature, it becomes critical to understand normal anatomy and locations with a high vasculature concentration surrounding the SI joint. METHODS The authors retrieved medical and radiographic records of patients who underwent computed tomography angiography (CTA) of the pelvis. Anterior and posterior compartments of the SI joint were established on the transverse view by creating an even coronal division of the SI joint. The superior, middle, and inferior compartments were established on the coronal view as three equal transverse compartments. The compartments in which vasculature was visualized were recorded. RESULTS Distribution of vasculature around the right and left hemipelvis concentrated in the inferior compartments and decreased in concentration while moving superiorly. Anterior compartments contain a higher vascularity than posterior compartments. Vasculature was present in <3% of the posterior middle, and posterior superior compartments while present in >83% of the inferior compartments. There were no significant differences with respect to vascular distribution when comparing the laterality of the right versus left hemipelvis. There were statistically significant relationships between vascular distribution and sex (P<0.05), as well as across self-reported ethnicity (P<0.05). CONCLUSIONS SI screw placement in the posterior superior has the lowest risk of iatrogenic vascular injury. Careful consideration should be taken during SI joint fusion surgery in the inferior compartments due to its high vasculature density.
Collapse
Affiliation(s)
- Zachary Brandt
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Kai Nguyen
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Gideon Harianja
- Department of Orthopedic Surgery, Loma Linda University Medical Center, Loma Linda, CA
| | - Kirulus Amin
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Adel Battikha
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Ninous Betdashtoo
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Rohan Kubba
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - David Shin
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Mark Oliinik
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Jacob Razzouk
- Department of Orthopaedic Surgery, Loma Linda University School of Medicine, Loma Linda University Medical Center, Loma Linda, CA
| | - Jun Ho Chung
- Department of Orthopedic Surgery, Loma Linda University Medical Center, Loma Linda, CA
| | - Olumide Danisa
- Department of Orthopedic Surgery, Loma Linda University Medical Center, Loma Linda, CA
| | - Wayne Cheng
- Department of Orthopedic Surgery, Jerry L. Pettis Veterans Affairs (VA) Medical Center, Loma Linda, CA
| |
Collapse
|
12
|
Jacobs K, Langenbach GEJ, Docter D, Cordewener PAM, van de Beek BJ, Korfage JAM, Visser SC, Peters JJ, Hagoort J, Lobbezoo F, de Bakker BS. Imaging the development of the human craniofacial arterial system - an experimental study. Pediatr Radiol 2025; 55:721-732. [PMID: 39254856 PMCID: PMC11982102 DOI: 10.1007/s00247-024-06044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The process of vascular development is essential for shaping complex craniofacial structures. Investigating the interplay between vascular development and orofacial morphogenesis holds critical importance in clinical practice and contributes to advancing our comprehension of (vascular) developmental biology. New insights into specific vascular developmental pathways will have far-reaching implications across various medical disciplines, enhancing clinical understanding, refining surgical techniques, and elucidating the origins of congenital abnormalities. Embryonic development of the craniofacial vasculature remains, however, under-exposed in the current literature. We imaged and created 3-dimensional (D) reconstructed images of the craniofacial arterial system from two early-stage human embryonic samples. OBJECTIVE The aim of this study was to investigate the vascular development of the craniofacial region in early-stage human embryos, with a focus on understanding the interplay between vascular development and orofacial morphogenesis. MATERIALS AND METHODS Reconstructions (3-D) were generated from high-resolution diffusible iodine-based contrast-enhanced computed tomography (diceCT) images, enabling visualization of the orofacial arterial system in human embryonic samples of Carnegie stages (CS) 14 and 18 from the Dutch Fetal Biobank, corresponding to weeks 7 and 8.5 of gestation. RESULTS From two human embryonic samples (ages CS 14 and 18), the vascular development of the orofacial region at two different stages of development was successfully stained with B-Lugol and imaged using a micro-computed tomography (micro-CT) scanner with resolutions of 2.5-μm and 9-μm voxel sizes, respectively. Additionally, educational 3-D reconstructions of the orofacial vascular system were generated using AMIRA 2021.2 software. CONCLUSION Micro-CT imaging is an effective strategy for high-resolution visualization of vascular development of the orofacial region in human embryonic samples. The generated interactive 3-D educational models facilitate better understanding of the development of orofacial structures.
Collapse
Affiliation(s)
- K Jacobs
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands.
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - G E J Langenbach
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands
| | - D Docter
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Pediatric Surgery, Emma Children's Hospital, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, The Netherlands
| | - P A M Cordewener
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands
| | - B J van de Beek
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands
| | - J A M Korfage
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands
| | - S C Visser
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, The Netherlands
| | - J J Peters
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, The Netherlands
| | - J Hagoort
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands
| | - F Lobbezoo
- Department of Oral Pain and Disfunction, Section Orofacial Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Gustav Mahlerlaan 3004, 1081LA, Amsterdam, The Netherlands
| | - B S de Bakker
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC, location AMC, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Pediatric Surgery, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Dr. Molewaterplein 40, Rotterdam, The Netherlands
| |
Collapse
|
13
|
March A, Hebner TS, Choe R, Benoit DSW. Leveraging the predictive power of a 3D in vitro vascularization screening assay for hydrogel-based tissue-engineered periosteum allograft healing. BIOMATERIALS ADVANCES 2025; 169:214187. [PMID: 39827700 PMCID: PMC11815559 DOI: 10.1016/j.bioadv.2025.214187] [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: 11/13/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
A common strategy for promoting bone allograft healing is the design of tissue-engineered periosteum (TEP) to orchestrate host-tissue infiltration. However, evaluating requires costly and time-consuming in vivo studies. Therefore, in vitro assays are necessary to expedite TEP designs. Since angiogenesis is a critical process orchestrated by the periosteum, this study investigates in vitro 3D cell spheroid vascularization as a predictive tool for TEP-mediated in vivo healing. Spheroids of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) are encapsulated in enzymatically-degradable poly (ethylene glycol)-based hydrogels and sprout formation, network formation, and angiogenic growth factor secretion are quantified. Hydrogels are also evaluated as TEP-modified allografts for in vivo bone healing with graft vascularization, callus formation, and biomechanical strength quantified as healing metrics. Evaluation of hydrogels highlights the importance of degradation, with 24-fold greater day 1 sprouts observed in degradable hydrogels in vitro and 4-fold greater graft-localized vascular volume at 6-weeks in vivo compared to non-degradable hydrogels. Correlations between in vitro and in vivo studies elucidate linear relationships when comparing in vitro sprout formation and angiocrine production with 3- and 6-week in vivo graft vascularization, 3-week cartilage callus, and 6-week bone callus, with a Pearson's R2 value equal to 0.97 for the linear correlation between in vitro sprout formation and 6-week in vivo vascular volume. Non-linear relationships are found between in vitro measures and bone torque strength at week 6. These correlations suggest that the in vitro sprouting assay has predictive power for in vivo vascularization and bone allograft healing.
Collapse
Affiliation(s)
- Alyson March
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA
| | - Tayler S Hebner
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA; Davidson School of Chemical Engineering, Purdue University, 480 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Regine Choe
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA; Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
14
|
Paslı B, Ülkir M, Günenç Beşer C. Morphometric, morphologic and topographic evaluation of diaphyseal nutrient foramina of the femur. Surg Radiol Anat 2025; 47:102. [PMID: 40131503 DOI: 10.1007/s00276-025-03611-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE Vascularization of bones is crucial for bone growth and repair. The nutrient artery, passing through the nutrient foramen, is key to bone blood supply, but its impact on fracture healing and complications is not fully understood. The study aims to investigate the morphology and location of the nutrient foramen in the femoral diaphysis and to understand its clinical implications for fractures. METHODS In this study, 88 adult dry femurs of unknown age and sex were examined. The characteristics of the nutrient foramina, including number, size, direction, and localization were evaluated. The foraminal index [(distance from foramen to proximal end of femur/total length of femur) × 100], was employed to categorize the regions [Region-I, 0-33.33; Region-II, 33.34-66.66; Region-III 66.67-100]. RESULTS The majority of the femurs had one or two foramina (92.94%). Of the total number of nutrient foramina, 121 (97.58%) were directed towards the proximal end, while three (2.42%) were horizontal. The majority of foramina were detected in sizes 18G (34.67%) and 20G (27.42%). All nutrient foramina were found on the posterior surface of the femur and adjacent to linea aspera. Sixteen nutrient foramina were located (12.90%) in Region-I, 104 (83.87%) in Region-II, and 4 (3.23%) in Region-III. CONCLUSIONS The nutrient foramina were typically located in the middle third of femur, adjacent to linea aspera on the posterior surface of femur. This observation indicates that the anterior surface is safer for surgery, while caution is needed near the linea aspera on the posterior surface.
Collapse
Affiliation(s)
- Bahattin Paslı
- Department of Anatomy, Hacettepe University School of Medicine, Sıhhiye, Çankaya, Ankara, 06100, Turkey.
| | - Mehmet Ülkir
- Department of Anatomy, Hacettepe University School of Medicine, Sıhhiye, Çankaya, Ankara, 06100, Turkey
| | - Ceren Günenç Beşer
- Department of Anatomy, Hacettepe University School of Medicine, Sıhhiye, Çankaya, Ankara, 06100, Turkey
| |
Collapse
|
15
|
Zhu M, Zhang H, Zhou Q, Sheng S, Gao Q, Geng Z, Chen X, Lai Y, Jing Y, Xu K, Bai L, Wang G, Wang J, Jiang Y, Su J. Dynamic GelMA/DNA Dual-Network Hydrogels Promote Woven Bone Organoid Formation and Enhance Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501254. [PMID: 40123197 DOI: 10.1002/adma.202501254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Bone organoids, in vitro models mimicking native bone structure and function, rely on 3D stem cell culture for self-organization, differentiation, ECM secretion, and biomineralization, ultimately forming mineralized collagen hierarchies. However, their development is often limited by the lack of suitable matrices with optimal mechanical properties for sustained cell growth and differentiation. To address this, a dynamic DNA/Gelatin methacryloyl (GelMA) hydrogel (CGDE) is developed to recapitulate key biochemical and mechanical features of the bone ECM, providing a supportive microenvironment for bone organoid formation. This dual-network hydrogel is engineered through hydrogen bonding between DNA and GelMA, combined with GelMA network crosslinking, resulting in appropriate mechanical strength and enhanced viscoelasticity. During a 21-day 3D culture, the CGDE hydrogel facilitates cellular migration and self-organization, promoting woven bone organoid (WBO) formation via intramembranous ossification. These WBOs exhibit spatiotemporal architectures supporting dynamic mineralization and tissue remodeling. In vivo studies demonstrate that CGDE-derived WBOs exhibit self-adaptive properties, enabling rapid osseointegration within 4 weeks. This work highlights the CGDE hydrogel as a robust and scalable platform for bone organoid development, offering new insights into bone biology and innovative strategies for bone tissue regeneration.
Collapse
Affiliation(s)
- Mengru Zhu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Hao Zhang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Qirong Zhou
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Qianmin Gao
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Zhen Geng
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Yingying Jing
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Ke Xu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Jianhua Wang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yingying Jiang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
16
|
Zhang H, Chang X, Liu X, Zhang B, Wang R, Wang Y, Dai S, Yao T, Zhang Q. Silencing of LOX-1 attenuates high glucose-induced ferroptosis in THVECs via the HIF-1α/SLC7A11 signaling pathway. Exp Cell Res 2025; 446:114451. [PMID: 40015503 DOI: 10.1016/j.yexcr.2025.114451] [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] [Revised: 01/11/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES Diabetic osteoporosis (DOP) represents a significant and serious complication associated with diabetes, characterized by a complex and inadequately understood pathophysiological mechanism. Recent studies have highlighted a robust association between DOP and ferroptosis. H-type vessels play a critical role in osteoporosis, while lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is associated with endothelial dysfunction related to diabetes. In this study, we investigate how LOX-1 affects ferroptosis in H-type vascular endothelial cells (THVECs) under high glucose (HG) conditions, aiming to elucidate the molecular mechanisms involved. METHODS THVECs were isolated from rats employing an enzymatic digestion method and subsequently validated through immunofluorescence analysis. The silencing of LOX-1 was achieved via transfection with a lentiviral vector. Cell viability was assessed using the CCK-8 assay, and ROS, MMP, GSH, MDA, and Fe2+ levels were assessed utilizing specific commercial kits. Western blotting and PCR assessed LOX-1, HIF-1α, SLC7A11, and GPX4 expression levels. RESULTS In high glucose conditions, LOX-1 expression at both protein and mRNA levels increased, while ROS, MDA, and Fe2+ rose, and MMP and GSH levels fell, resulting in ferroptosis in THVECs. This condition could be reversed by silencing LOX-1 or by administering the ferroptosis inhibitor (Fer-1). Further analysis showed that silencing LOX-1 enhanced the expression of HIF-1α, SLC7A11, and GPX4, which mitigated ferroptosis in THVECs. CONCLUSIONS Downregulation of LOX-1 alleviates high glucose-induced ferroptosis in THVECs via the HIF-1α/SLC7A11 pathway. This suggests that LOX-1 functions as a critical target for regulating ferroptosis in THVECs, providing a novel insight into the pathological mechanisms associated with DOP.
Collapse
Affiliation(s)
- Haiqi Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinying Chang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xuan Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Baozhuan Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Rongrong Wang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yuhui Wang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Simeng Dai
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Tonghan Yao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Qi Zhang
- Department of Gerontology, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
17
|
Kotsifaki A, Kalouda G, Maroulaki S, Foukas A, Armakolas A. The Genetic and Biological Basis of Pseudoarthrosis in Fractures: Current Understanding and Future Directions. Diseases 2025; 13:75. [PMID: 40136615 PMCID: PMC11941250 DOI: 10.3390/diseases13030075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Pseudoarthrosis-the failure of normal fracture healing-remains a significant orthopedic challenge affecting approximately 10-15% of long bone fractures, and is associated with significant pain, prolonged disability, and repeated surgical interventions. Despite extensive research into the pathophysiological mechanisms of bone healing, diagnostic approaches remain reliant on clinical findings and radiographic evaluations, with little innovation in tools to predict or diagnose non-union. The present review evaluates the current understanding of the genetic and biological basis of pseudoarthrosis and highlights future research directions. Recent studies have highlighted the potential of specific molecules and genetic markers to serve as predictors of unsuccessful fracture healing. Alterations in mesenchymal stromal cell (MSC) function, including diminished osteogenic potential and increased cellular senescence, are central to pseudoarthrosis pathogenesis. Molecular analyses reveal suppressed bone morphogenetic protein (BMP) signaling and elevated levels of its inhibitors, such as Noggin and Gremlin, which impair bone regeneration. Genetic studies have uncovered polymorphisms in BMP, matrix metalloproteinase (MMP), and Wnt signaling pathways, suggesting a genetic predisposition to non-union. Additionally, the biological differences between atrophic and hypertrophic pseudoarthrosis, including variations in vascularity and inflammatory responses, emphasize the need for targeted approaches to management. Emerging biomarkers, such as circulating microRNAs (miRNAs), cytokine profiles, blood-derived MSCs, and other markers (B7-1 and PlGF-1), have the potential to contribute to early detection of at-risk patients and personalized therapeutic approaches. Advancing our understanding of the genetic and biological underpinnings of pseudoarthrosis is essential for the development of innovative diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Amalia Kotsifaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Georgia Kalouda
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Sousanna Maroulaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Athanasios Foukas
- Third Department of Orthopaedic Surgery, “KAT” General Hospital of Athens, 2, Nikis Street, 14561 Kifissia, Greece;
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| |
Collapse
|
18
|
Roshini N, Yuvaraj MF, Kasirajan SP, Karunakaran B, Govindan L, Caleb JTD, Sivalingam AM, Siva T, Kumar S. Nutrient foramina of human fibula: morphometric analysis and clinical relevance. J Bone Miner Metab 2025; 43:149-157. [PMID: 39623083 DOI: 10.1007/s00774-024-01568-6] [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: 09/15/2023] [Accepted: 11/17/2024] [Indexed: 04/13/2025]
Abstract
BACKGROUND The fibula, situated laterally in the leg, receives vital nutrition through nutrient arteries during embryonic bone growth and early ossification. This study aims to assess the direction, distance, location, number, and foraminal index of nutrient foramina in dry fibulae from the South Indian population. MATERIALS AND METHODS A descriptive cross-sectional analysis involved 63 dry adult human fibulae sourced from the Department of Anatomy, Saveetha Medical College and Hospital, Thandalam. Parameters like fibula length, location, number, and direction of vascular foramina were recorded. Statistical analyses were performed on morphometric data and foraminal index. RESULTS The mean fibula length was 34.68 ± 2.11 cm. Among the fibulae, 88.88% had a single nutrient foramen, 4.76% had dual foramina, and 6.34% lacked nutrient foramina. Most single foramina were found on the medial crest (66.66%), followed by between the medial crest and posterior border (20.63%). Nutrient foramina were primarily located in Zone II (87.30%), followed by Zone III (11.11%) and Zone I (1.58%). Directionally, 85.71% pointed downward, while 14.28% pointed upward. The mean foraminal index was 40.85 ± 6.78, ranging from 32.57 to 56.25. CONCLUSION Zone II, particularly on the medial crest, was the most prevalent location for vascular foramina in the fibula. Dual foramina occurred in 6.34% of cases. This precise anatomical knowledge is valuable for various medical professionals, including anthropologists, forensic experts, radiologists, plastic surgeons, and orthopedic surgeons, especially in procedures involving vascularized fibular bone grafts.
Collapse
Affiliation(s)
- N Roshini
- Department of Anatomy, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, 600116, India
| | - Maria Francis Yuvaraj
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 602105, India.
| | - Sankaran Ponnusamy Kasirajan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, 522503, India
| | - Balaji Karunakaran
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 602105, India
| | - Lakshmanan Govindan
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 602105, India
| | - John T D Caleb
- Department of Anatomy, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 602105, India
| | - Azhagu Madhavan Sivalingam
- Natural Products & Nanobiotechnology Research Lab, Department of Community Medicine, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, Chennai, Tamil Nadu, 602 105, India.
| | - T Siva
- Department of Anatomy, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, 600116, India
| | - Sathish Kumar
- Department of Community Medicine, Vinayaka Mission's Medical College and Hospital, VMRF-DU, Karaikal, 609609, India
| |
Collapse
|
19
|
Xue P, Wang J, Fu Y, He H, Gan Q, Liu C. Material-Mediated Immunotherapy to Regulate Bone Aging and Promote Bone Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409886. [PMID: 39981851 DOI: 10.1002/smll.202409886] [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: 10/25/2024] [Revised: 01/19/2025] [Indexed: 02/22/2025]
Abstract
As the global population ages, an increasing number of elderly people are experiencing weakened bone regenerative capabilities, resulting in slower bone repair processes and associated risks of various complications. This review outlines the research progress on biomaterials that promote bone repair through immunotherapy. This review examines how manufacturing technologies such as 3D printing, electrospinning, and microfluidic technology contribute to enhancing the therapeutic effects of these biomaterials. Following this, it provides detailed introductions to various anti-osteoporosis drug delivery systems, such as injectable hydrogels, nanoparticles, and engineered exosomes, as well as bone tissue engineering materials and coatings used in immunomodulation. Moreover, it critically analyzes the current limitations of biomaterial-mediated bone immunotherapy and explores future research directions for material-mediated bone immunotherapy. This review aims to inspire new approaches and broaden perspectives in addressing the challenges of bone repair and aging by exploring innovative biomaterial-mediated immunotherapy strategies.
Collapse
Affiliation(s)
- Pengfei Xue
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics, Tongji University, Zhangwu Road 100, Shanghai, 200092, China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
20
|
Park H, Narayanan SA, Caldwell JT, Behnke BJ, Muller-Delp JM, Delp MD. Effects of aging and exercise training on bone and marrow blood flow and vascular function. Bone 2025; 192:117335. [PMID: 39561871 DOI: 10.1016/j.bone.2024.117335] [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: 08/09/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/21/2024]
Abstract
Aging leads to progressive bone loss, which is associated with impaired bone and marrow perfusion. The purpose of this study was to determine whether chronic exercise training enhances blood flow to the femur at rest and during exercise, and elucidate whether putative changes in training-induced bone perfusion are associated with alterations in the intrinsic vasomotor properties of the femoral principal nutrient artery (PNA) in old age. Young (4-6 mo old) and old (20-22 mo old) male Fischer-344 rats were either treadmill exercise trained (ET) or remained sedentary (SED). Regional blood flow to the femur was assessed at rest and during treadmill exercise. Endothelium-dependent (acetylcholine, ACh) and -independent (Dea-NONOate) vasodilator, and vasoconstrictor (phenylephrine (PE), KCl and myogenic) responses of femoral PNAs were determined. Exercise training led to higher blood flow to distal metaphysis and epiphysis in old rats at rest, and old ET rats showed greater regional blood flow during exercise compared to old SED rats. The increased blood flow to the proximal and distal metaphysis and epiphysis were also higher in old ET rats than that in young ET rats. Exercise training enhanced the vasodilator response to ACh, corresponding to increased eNOS expression in femoral PNAs from both young and old rats. Aging did not alter PE- or KCl-induced vasoconstriction, whereas myogenic responses were impaired. Exercise training enhanced vasoconstrictor responses to PE in old rats but had no effect on KCl or myogenic responses in either group. These data demonstrate that exercise training enhances both regional bone and marrow blood flow and vasodilator responses, which are impaired in the femora of old SED rats.
Collapse
Affiliation(s)
- Hyerim Park
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - S Anand Narayanan
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Jacob T Caldwell
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Bradley J Behnke
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Judy M Muller-Delp
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | - Michael D Delp
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
21
|
Lee E, Lialios P, Curtis M, Williams J, Kim Y, Salipante P, Hudson S, Esch MB, Levi M, Kitlinska J, Alimperti S. Glucocorticoids Alter Bone Microvascular Barrier via MAPK/Connexin43 Mechanisms. Adv Healthc Mater 2025; 14:e2404302. [PMID: 39831839 PMCID: PMC11912118 DOI: 10.1002/adhm.202404302] [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: 11/02/2024] [Revised: 12/30/2024] [Indexed: 01/22/2025]
Abstract
Glucocorticoids (GCs) are standard-of-care treatments for inflammatory and immune disorders, and their long-term use increases the risk of osteoporosis. Although GCs decrease bone functionality, their role in bone microvasculature is incompletely understood. Herein, the study investigates the mechanisms of bone microvascular barrier function via osteoblast-endothelial interactions in response to GCs. The animal data shows that prednisolone (Psl) downregulated the osteoblast function and microvessel number and size. To investigate the role of GCs in bone endothelial barrier function further, a bicellular microfluidic in vitro system is developed and utilized, which consists of three-dimensional (3D) perfusable microvascular structures embedded in collagen I/osteoblast matrix. Interestingly, it is demonstrated that GCs significantly inhibit osteogenesis and microvascular barrier function by interfering with endothelial-osteoblast interactions. This effect is triggered by MAPK-induced phosphorylation of connexin43 (Cx43) at Ser282. Collectively, this study sheds light on microvascular function in bone disorders, as osteoporosis, and permits to capture dynamic changes in endothelial-bone interactions under GCs by dissecting the MAPK/Cx43 mechanism and proposing this as a potential target for bone diseases.
Collapse
Affiliation(s)
- Eun‐Jin Lee
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Microsystems and Nanotechnology DivisionPhysical Measurement LaboratoryNational Institute of Standards and TechnologyGaithersburgMD20899USA
- Department of Chemistry and BiochemistryCollege of ComputerMathematical and Natural SciencesUniversity of MarylandCollege ParkMD20742USA
| | - Peter Lialios
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - Micaila Curtis
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - James Williams
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - Yoontae Kim
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - Paul Salipante
- Materials Science and Engineering DivisionMaterial Measurement LaboratoryNational Institute of Standards and TechnologyGaithersburgMD20899USA
| | - Steven Hudson
- Materials Science and Engineering DivisionMaterial Measurement LaboratoryNational Institute of Standards and TechnologyGaithersburgMD20899USA
| | - Mandy B. Esch
- Microsystems and Nanotechnology DivisionPhysical Measurement LaboratoryNational Institute of Standards and TechnologyGaithersburgMD20899USA
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
- Biological and Biomedical Engineering CenterSchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| | - Stella Alimperti
- Department of Biochemistry and Molecular and Cellular BiologySchool of MedicineGeorgetown UniversityWashingtonDC20057USA
| |
Collapse
|
22
|
Chen Q, Wang D, Shang J. Experimental research of different forms of autolyzed antigen-extracted allogeneic bone combined with vascular endothelial growth factor for the repair of bone defects. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2025; 126:102066. [PMID: 39245287 DOI: 10.1016/j.jormas.2024.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE To investigate the effect of different forms of autolyzed antigen-extracted allogeneic(AAA) bone combined with vascular endothelial growth factor (VEGF) on bone reconstruction. METHOD The AAA bone was made into a block and a granule shape, and mixed with VEGF to prepare VEGF bone. Establishment of rat calvarium defect animal model, it is divided into 5 groups. With block bone, granular bone, block VEGF bone, granular VEGF bone was implanted in the bone defect for repair as the experimental group. The defect area was evaluated by histological and CBCT analysis 4 weeks postoperatively. RESULTS Postoperative 4 weeks imaging results showed that there was no high-density shadow in the bone defect area of the blank group and the volume of high-density shadow in the bone defect area of the experimental group was different. Histological results showed that no osteoblasts were found in the blank group, and new bone was formed in the experimental group. The effect of bone formation in the granular bone was better than that in the block bone, and the amount of new bone formation in the VEGF bone group was higher than that of the single bone group. CONCLUSION Granular bone has a better osteogenesis effect than block bone. The effect of allogeneic bone combined with VEGF in promoting new bone formation in the area of the bone defect is better than that of allogeneic bone alone. These results provide a theoretical and practical basis for its further clinical application.
Collapse
Affiliation(s)
- Qiang Chen
- Department of the First Clinical Division, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.
| | - Dandan Wang
- Department of the First Clinical Division, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Jiaxin Shang
- Department of the First Clinical Division, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| |
Collapse
|
23
|
March A, Wu H, Choe R, Benoit DSW. Optimizing Tissue-Engineered Periosteum Biochemical Cues to Hasten Bone Allograft Healing. J Biomed Mater Res A 2025; 113:e37890. [PMID: 40033815 DOI: 10.1002/jbm.a.37890] [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: 12/20/2024] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Although allografts remain the gold standard for treating critical-size bone defects, ~60% fail within 10 years of implantation. To emulate periosteum-mediated healing of live autografts, we have developed a tissue-engineered periosteum (TEP) to improve allograft healing. The TEP comprises cell-degradable poly(ethylene glycol) hydrogels encapsulating mouse mesenchymal stem cells and osteoprogenitor cells to mimic the periosteal cell population. Despite improvements in allograft healing, several limitations were observed using the TEP, specifically the modulation of host tissue infiltration and remodeling to support graft-localized vascular volume and callus bridging. Therefore, hydrogel biochemical cues were incorporated into TEP to enable cell-matrix interactions and remodeling critical for tissue infiltration. Adhesive peptide functionalization (RGD, YIGSR, and GFOGER) and enzymatic degradation rate (GPQGIWGQ, IPESLRAG, and VPLSLYSG) were screened using an in vitro 3D cell spheroid assay and design of experiments (DOE) to identify hydrogels that best supported tissue infiltration and integration. DOE analysis of various adhesive peptide combinations was used to optimize functionalization, revealing that individual RGD-functionalization and GFOGER-functionalization maximized in vitro cell infiltration. RGD and GFOGER hydrogels were then investigated in vivo as TEP (RGD-TEP and GFOGER-TEP, respectively) to evaluate the effect of hydrogel functionalization on TEP-mediated allograft healing in a murine femur defect model. RGD- and GFOGER-TEP promoted bone graft healing, with both groups exhibiting a 1.9-fold increase in bone callus volume over unmodified allografts at 3 weeks post-implantation. RGD-TEP promoted more significant bone tissue development, but GFOGER-TEP promoted greater torsional biomechanics over time. The few differences observed between TEP groups suggest hydrogel functionalization has a limited effect on TEP-mediated healing, with cell delivery via the TEP enough to improve bone regeneration. Future studies aim to investigate additional adhesive peptides with diverse combinations to identify potential synergies between adhesive peptides to promote TEP-mediated bone allograft healing.
Collapse
Affiliation(s)
- Alyson March
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Hao Wu
- Institute of Optics, University of Rochester, Rochester, New York, USA
| | - Regine Choe
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Department of Electrical and Computer Engineering, University of Rochester, Rochester, New York, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon, USA
| |
Collapse
|
24
|
Gracea RS, Suryani IR, Fontenele RC, Gaêta-Araujo H, Radi S, Elgarba BM, Shujaat S, Coropciuc R, Jacobs R. Alveolar socket surface area as a local risk factor for MRONJ development in oncologic patients on polypharmacy. Clin Oral Investig 2025; 29:123. [PMID: 39920415 PMCID: PMC11805769 DOI: 10.1007/s00784-025-06200-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025]
Abstract
OBJECTIVES To determine the impact of alveolar socket surface area and number of root extractions for developing medication-related osteonecrosis of the jaw (MRONJ) in polypharmacy patients following multiple tooth extractions. MATERIALS AND METHODS A retrospective sample of 40 patients was recruited, including 20 polypharmacy patients (109 tooth extractions) who developed MRONJ in at least one of the extraction sites, matched with 20 controls (100 tooth extractions). Tooth-specific alveolar socket surface areas were assessed using CBCT scans from the control group to establish reference values for alveolar socket surface areas in polypharmacy patients with MRONJ. Correlations between the number of extracted tooth roots, alveolar socket surface area, and development of MRONJ were analysed within the polypharmacy group. RESULTS 40% of tooth extractions in polypharmacy patients undergoing multiple extractions resulted in the development of MRONJ, with a higher prevalence observed in the mandible (44%). Among the extracted mandibular tooth roots, half were susceptible to MRONJ, and 45% of the exposed socket surface area was affected. Both jaws showed an increased risk (20%) of MRONJ following molar extraction. A strong positive correlation existed between extraction sites that developed MRONJ and both the number of mandibular tooth roots extracted (r = + 0.861; p < 0.001) and the total exposed alveolar socket surface area (r = + 0.757; p < 0.001). However, no significant correlations were observed in the upper jaw. CONCLUSIONS This study is the first to demonstrate that both mandibular alveolar socket surface area and number of extracted tooth roots are positively related to extraction sites developing MRONJ in polypharmacy patients undergoing multiple tooth extractions. CLINICAL RELEVANCE Identifying high-risk patients and implementing preventive strategies can reduce MRONJ incidence, underscoring the need for careful management of polypharmacy patients, especially those undergoing mandibular tooth extractions.
Collapse
Affiliation(s)
- Rellyca Sola Gracea
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia.
- OMFS-Impath Research Group, KU Leuven, Kapucijnenvoer 7, Leuven, B-3000, Belgium.
| | - Isti Rahayu Suryani
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rocharles Cavalcante Fontenele
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Hugo Gaêta-Araujo
- Department of Stomatology, Public Health and Forensic Dentistry, Division of Oral Radiology, School of Dentistry of Ribeirão Preto, University of São Paulo, São Paulo, Ribeirão Preto, Brazil
| | - Sonya Radi
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Bahaaeldeen M Elgarba
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Prosthodontics, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Sohaib Shujaat
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
- King Abdullah International Medical Research Center, Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Ruxandra Coropciuc
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Reinhilde Jacobs
- OMFS-IMPATH Research Group, Department of Imaging & Pathology, Faculty of Medicine, KU Leuven & Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.
- Department of Dental Medicine, Karolinska Institutet, Alfred Nobels Allé 8, Huddinge, Stockholm, 141 50, Sweden.
| |
Collapse
|
25
|
Shao B, Fu Y, Li B, Huo S, Du J, Zhang X, Yin X, Li Y, Cao Z, Song M. Icariin-loaded chitosan/β-glycerophosphate thermosensitive hydrogel enhanced infection control and bone regeneration in canine with infectious bone defects. J Biomater Appl 2025; 39:696-713. [PMID: 39411862 DOI: 10.1177/08853282241288323] [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: 01/29/2025]
Abstract
Faced with infectious bone defects, the development of a thermosensitive hydrogel containing icariin (ICA) represents a promising therapeutic strategy targeting infection control and bone regeneration. In this study, we prepared and evaluated the physicochemical properties, in vitro and in vivo drug release, antimicrobial activity, anti-inflammatory properties, and bone repair effects of ICA/Chitosan/β-Glycerophosphate (ICA/CTS/β-GP) thermosensitive hydrogel. Our findings demonstrate that the ICA/CTS/β-GP thermosensitive hydrogel undergoes a liquid-to-gel transition at body temperature, which is crucial for maintaining local drug release at the defect site. Additionally, the hydrogel exhibited sustained release of ICA over 28 days, showing high antimicrobial activity against Staphylococcus aureus and good biocompatibility in blood compatibility tests. In a canine model of infectious bone defects, the ICA/CTS/β-GP thermosensitive hydrogel showed effective infection control and modulated inflammation, vascular formation, and bone factor expression, while also activating the Wnt/β-catenin signaling pathway. In conclusion, the ICA/CTS/β-GP thermosensitive hydrogel could control infection and repair bone tissue. Its antimicrobial and osteogenic properties provide hope for its clinical application.
Collapse
Affiliation(s)
- Bing Shao
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Northeast Agricultural University Animal Hospital Co.Ltd, Harbin, China
| | - Yang Fu
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Department of Veterinary Medicine, Heze Vocational College, Heze, China
| | - Bo Li
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Siming Huo
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jiayu Du
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuliang Zhang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zheng Cao
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Miao Song
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
26
|
Cheng Z, Wang Y, Lin H, Chen Z, Qin R, Wang T, Xu H, Du Y, Yuan H, Pan Y, Jiang H, Jiang X, Jiang J, Wu F, Wang Y. Engineering Dual Active Sites and Defect Structure in Nanozymes to Reprogram Jawbone Microenvironment for Osteoradionecrosis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413215. [PMID: 39686746 PMCID: PMC11809426 DOI: 10.1002/advs.202413215] [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: 10/21/2024] [Revised: 11/26/2024] [Indexed: 12/18/2024]
Abstract
Four to eight percent of patients with head and neck cancer will develop osteoradionecrosis of the jaw (ORNJ) after radiotherapy. Various radiation-induced tissue injuries are associated with reactive oxygen and nitrogen species (RONS) overproduction. Herein, Fe doping is used in VOx (Fe-VOx) nanozymes with multienzyme activities for ORNJ treatment via RONS scavenging. Fe doping can induce structure reconstruction of nanozymes with abundant defect production, including Fe substitution and oxygen vacancies (OVs), which markedly increased multiple enzyme-mimicking activity. Catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) enzyme-like performance of Fe-VOx can effectively reprogram jawbone microenvironment to restore mitochondrial dysfunction and enhance mitophagy. Moreover, the surface plasmon resonance (SPR) effect of Fe-VOx made it a good photothermal nanoagents for inhibiting jaw infection. Thus, this work demonstrated that Fe-VOx nanozymes can efficiently scavenge RONS, activate mitophagy, and inhibit bacteria, which is potential for ORNJ treatment.
Collapse
Affiliation(s)
- Zheng Cheng
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Yuchen Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Haobo Lin
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Ziyu Chen
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| | - Ran Qin
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Tianxiao Wang
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| | - Hang Xu
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200120China
| | - Yifei Du
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Hua Yuan
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Yongchu Pan
- Department of OrthodonticThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
| | - Huijun Jiang
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| | - Xinquan Jiang
- Department of ProsthodonticsShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai Engineering Research Center of Advanced Dental Technology and MaterialsNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyNo. 639 Zhizaoju RoadShanghai200011China
| | - Jiandong Jiang
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Fan Wu
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| | - Yuli Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityState Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral DiseasesJiangsu Province Engineering Research Centre of Stomatological Translational MedicineNanjing Medical UniversityNanjingJiangsu210029China
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular DiseasesMinistry of EducationInternational Joint Laboratory for Drug Target of Critical IllnessesSchool of PharmacyNanjing Medical UniversityNanjingJiangsu211166China
| |
Collapse
|
27
|
Shah MAA, Tang W, Zhang JH, Chen C, Wang JW, Lü SJ, Yu XT, Zhang ZJ, Li C, Yu SB, Sui HJ. Microvasculature and trabecular bone in beagle proximal femur: Microstructural insights. Ann Anat 2025; 258:152368. [PMID: 39643064 DOI: 10.1016/j.aanat.2024.152368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Avascular necrosis of femoral head and malunion are frequent post-operative complications of femoral neck fractures. To optimize surgical techniques, this study aims to provide a microstructural understanding of intraosseous microvasculature and the trabecular bone of the femoral head and neck. STUDY DESIGN This anatomical study analyzed twenty-eight femora from fourteen cadaveric beagles. Common iliac arteries were infused with colored silicone-resin for vascular visualization, followed by non-decalcified hard tissue processing using the EXAKT®, and Masson's trichrome staining. Morphology and histomorphometric analysis were performed by Nikon NIS Elements BR and ImageJ-fiji. RESULTS Histomorphometry revealed thin, elongated trabeculae with high vascularity aligned parallel in the neck; numerous intraosseous anastomoses at the neck-shaft and head-neck junctions; thick trabeculae with smaller marrow cavities, and dense branching vascular networks near the cortex in the head. Quantitative analysis showed an inverse correlation between trabecular density and mean vascular density/vascular length density, with no significant sex or side differences. Dense connective tissue fibers maintained the microvasculature and trabeculae structure. CONCLUSION The femoral neck displayed an outside-in microvascular pattern via retinacular branches. Conversely, the femoral head had an inside-out pattern through epiphyseal branches reinforced by medullary branches. Dense intraosseous microvasculature aligned sub-cortically. The study identified a potential anatomical safe zone for screwing in femoral neck fractures in beagles. These findings provide an anatomical basis for translational research in joint preservation techniques for humans.
Collapse
Affiliation(s)
- M Adeel Alam Shah
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| | - Wei Tang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Jing-Hui Zhang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Cheng Chen
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Jia-Wei Wang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Shu-Jun Lü
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Xin-Tong Yu
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Zhi-Jun Zhang
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Chan Li
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Sheng-Bo Yu
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| | - Hong-Jin Sui
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
28
|
Müller D, Klotsche J, Kosik MB, Perka C, Buttgereit F, Hoff P, Gaber T. Fracture Fusion on Fast-Forward: Locally Administered Deferoxamine Significantly Enhances Fracture Healing in Animal Models: A Systematic Review and Meta-Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413290. [PMID: 39840407 PMCID: PMC11848589 DOI: 10.1002/advs.202413290] [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: 10/20/2024] [Revised: 12/08/2024] [Indexed: 01/23/2025]
Abstract
Fractures, with a yearly incidence of 1.2%, can lead to healing complications in up to 10% of cases. The angiogenic stimulant deferoxamine (DFO) is recognized for enhancing bone healing when administered into the fracture gap. This systematic review with meta-analysis investigates the effect of local DFO application on bone healing in rat and mouse models. EMBASE, MEDLINE (PubMed), and Web of Science are systematically searched in January 2024. The study is prospectively registered in PROSPERO (CRD42024492533), and the SYRCLE tool is used to assess study quality and risk of bias. Outcome values contain the primary endpoint bone volume fraction (BV/TV) as well as the secondary endpoints bone volume, tissue volume, bone mineral density, trabecular separation, trabecular thickness, vessel formation and the mechanical properties, assessed by µCT, angiography and mechanical strength tests. Out of 21 included studies, 18 qualify for meta-analysis, involving 539 animals. DFO-treated groups exhibit significantly higher BV/TV values (p < 0.0001) compared to controls, with similarly significant improvements in secondary outcomes. These findings highlight the substantial benefit of DFO in promoting bone healing, especially after radiotherapy. Rapid clinical implementation is recommended to help patients at high risk of fracture healing complications.
Collapse
Affiliation(s)
- Daniel Müller
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
| | - Jens Klotsche
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| | - Magdalena B. Kosik
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
| | - Carsten Perka
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinCharitéCenter for Orthopedics und Traumatology10117BerlinGermany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| | - Paula Hoff
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- MVZ Endokrinologikum Berlin am Gendarmenmarkt10117BerlinGermany
| | - Timo Gaber
- Department of Rheumatology and Clinical ImmunologyCharité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin10117BerlinGermany
- Deutsches Rheumaforschungszentrum Berlin (DRFZ)a Leibniz Institute10117BerlinGermany
| |
Collapse
|
29
|
Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H, Xu Q. Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater 2025; 44:283-318. [PMID: 39507371 PMCID: PMC11539077 DOI: 10.1016/j.bioactmat.2024.10.017] [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: 04/06/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Regenerative medicine endeavors to restore damaged tissues and organs utilizing biological approaches. Utilizing biomaterials to target and regulate the pathophysiological processes of injured tissues stands as a crucial method in propelling this field forward. The Extracellular Vesicles-in-Hydrogel (EViH) system amalgamates the advantages of extracellular vesicles (EVs) and hydrogels, rendering it a prominent biomaterial in regenerative medicine with substantial potential for clinical translation. This review elucidates the development and benefits of the EViH system in tissue regeneration, emphasizing the interaction and impact of EVs and hydrogels. Furthermore, it succinctly outlines the pathophysiological characteristics of various types of tissue injuries such as wounds, bone and cartilage injuries, cardiovascular diseases, nerve injuries, as well as liver and kidney injuries, underscoring how EViH systems target these processes to address related tissue damage. Lastly, it explores the challenges and prospects in further advancing EViH-based tissue regeneration, aiming to impart a comprehensive understanding of EViH. The objective is to furnish a thorough overview of EViH in enhancing regenerative medicine applications and to inspire researchers to devise innovative tissue engineering materials for regenerative medicine.
Collapse
Affiliation(s)
- Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yansheng Shan
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| |
Collapse
|
30
|
El Kandoussi A, Staffa SJ, Ömeroğlu E, Hung YP, Bauer F, Lozano-Calderon S, Chang CY. Triangular Margin: Reliable Imaging Feature of Fibrous Dysplasia in Long Bones? J Comput Assist Tomogr 2025:00004728-990000000-00423. [PMID: 39876555 DOI: 10.1097/rct.0000000000001731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025]
Abstract
OBJECTIVE To determine the utility of a triangular margin as an imaging diagnostic feature for fibrous dysplasia. MATERIALS AND METHODS We retrospectively reviewed all surgically biopsied or managed benign and malignant bone tumors by a single orthopedic oncologist over 19 years (2003 to 2022). A musculoskeletal radiologist and an orthopedic oncologist, both with >10 years of experience, retrospectively evaluated all imaging in consensus. Groups were compared using the χ2 test. RESULTS There were a total of 152 subjects [mean age 49±21 (range 7.8 to 91) years]; 80 (53%) females and 72 (47%) males. There were 52 subjects with fibrous dysplasia, 31 subjects with other benign bone tumors, and 69 subjects with malignant bone tumors. The sensitivity and specificity of a triangular margin for distinguishing fibrous dysplasia from other benign or malignant bone tumors were 74% and 96% on radiographs, 73% and 100% on CT, and 78% and 91% on MRI, respectively. The triangular margin was more prevalent in fibrous dysplasia (85%) versus benign (16%) and malignant (1.6%) primary bone tumors in all 3 modalities (P<0.001). Multivariate analysis of the aggregated imaging data suggests that if a lesion has a triangular margin, it is 14 times more likely to be a fibrous dysplasia than another benign bone tumor (P=0.012). CONCLUSIONS The presence of a triangular margin could increase a radiologist's confidence that a bone tumor is fibrous dysplasia.
Collapse
Affiliation(s)
- Amine El Kandoussi
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital, Boston, MA
| | - Steven J Staffa
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA
| | - Emre Ömeroğlu
- Department of Radiology, MetroWest Medical Center, Worcester, MA
| | | | - Fabian Bauer
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital, Boston, MA
| | | | - Connie Y Chang
- Department of Radiology, Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
31
|
Feng J, Zhu C, Zou J, Zhang L. Hyperbaric Oxygen Therapy for the Treatment of Bone-Related Diseases. Int J Mol Sci 2025; 26:1067. [PMID: 39940834 PMCID: PMC11817436 DOI: 10.3390/ijms26031067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Hyperbaric oxygen therapy (HBOT) is a therapeutic modality that enhances tissue oxygenation by delivering 100% oxygen at pressures greater than 1 absolute atmosphere. In recent years, HBOT has shown considerable potential in the treatment of bone diseases. While excess oxygen was once thought to induce oxidative stress, recent studies indicate that when administered within safe limits, HBOT can notably promote bone healing and repair. Extensive basic research has demonstrated that HBOT can stimulate the proliferation and differentiation of osteoblasts and encourage bone angiogenesis. Furthermore, HBOT has been shown to exert a beneficial influence on bone metabolism by modulating the inflammatory response and redox status. These mechanisms are closely related to core issues of bone biology. Specifically, in the context of fracture healing, bone defect repair, and conditions such as osteoporosis, HBOT targets the key bone signaling pathways involved in bone health, thereby exerting a therapeutic effect. Several clinical studies have demonstrated the efficacy of HBOT in improving bone health. However, the optimal HBOT regimen for treating various bone diseases still requires further definition to expand the indications for its clinical application. This paper outlines the mechanisms of HBOT, focusing on its antioxidant stress, promotion of bone vascularization, and anti-inflammatory properties. The paper also describes the application of HBOT in orthopedic diseases, thereby providing a scientific basis for the development of precise and personalized HBOT treatment regimens in clinical orthopedics.
Collapse
Affiliation(s)
- Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.F.); (C.Z.); (J.Z.)
| | - Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.F.); (C.Z.); (J.Z.)
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.F.); (C.Z.); (J.Z.)
| | - Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
32
|
Chen F, Wang P, Dai F, Zhang Q, Ying R, Ai L, Chen Y. Correlation Between Blood Glucose Fluctuations and Osteoporosis in Type 2 Diabetes Mellitus. Int J Endocrinol 2025; 2025:8889420. [PMID: 39949568 PMCID: PMC11824305 DOI: 10.1155/ije/8889420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/08/2025] [Indexed: 02/16/2025] Open
Abstract
The purpose of this review is to investigate the impacts of blood glucose fluctuations on diabetic osteoporosis, a complication of Type 2 diabetes mellitus (T2DM) that remains poorly understood. We reviewed the current evidence of the relationship between blood glucose fluctuations and diabetic osteoporosis in patients with T2DM. The findings indicate that blood glucose fluctuations may contribute to inhibiting the processes of bone formation and resorption, promoting diabetic osteoporosis and fractures in T2DM. Mechanistic studies, both in vitro and in vivo, reveal that these effects are largely mediated by oxidative stress, advanced glycation end products, inflammatory mediators, and multiple pathways inducing cell apoptosis or autophagy. Thus, maintaining the long-term stability of blood glucose levels emerges as a target to be pursued in clinical practice in order to safely reduce mean blood glucose and for its direct effects on osteoporosis and fractures in T2DM.
Collapse
Affiliation(s)
- Fuhua Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ping Wang
- Department of Endocrinology, The 2nd People's Hospital of Anhui, Hefei, Anhui, China
| | - Fang Dai
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ruixue Ying
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Liya Ai
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiqing Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
33
|
Shen C, Shen A. 4D printing: innovative solutions and technological advances in orthopedic repair and reconstruction, personalized treatment and drug delivery. Biomed Eng Online 2025; 24:5. [PMID: 39838448 PMCID: PMC11748259 DOI: 10.1186/s12938-025-01334-3] [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: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
With precise control of smart materials deformation in time dimension, doctors can customize orthopedic implants. This review focuses on the advances of 4D printing technology in orthopedics, including its applications in bone repair and reconstruction, personalized treatment, and drug delivery. 4D printing enables the creation of bionic scaffolds and fixation devices for bone repair, customized implants matching patients' conditions for personalized treatment, and specific carriers for accurate drug release and delivery, which together contribute to accelerating bone healing, providing exclusive treatments, enhancing therapeutic effects and reducing side effects, thus helping improve orthopedic medicine. It offers comprehensive reference materials for relevant medical personnel.
Collapse
Affiliation(s)
- Chenxi Shen
- Chongqing Medical University, 61 University Town Middle RoadShapingba District, Chongqing, 400000, People's Republic of China.
| | - Aiyong Shen
- The Fourth People's Hospital of Wujiang District, Suzhou, 215231, Jiangsu Province, People's Republic of China
| |
Collapse
|
34
|
Zhou X, Chen S, Pich A, He C. Advanced Bioresponsive Drug Delivery Systems for Promoting Diabetic Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:182-207. [PMID: 39666445 DOI: 10.1021/acsbiomaterials.4c02037] [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: 12/14/2024]
Abstract
The treatment of bone defects in diabetes mellitus (DM) patients remains a major challenge since the diabetic microenvironments significantly impede bone regeneration. Many abnormal factors including hyperglycemia, elevated oxidative stress, increased inflammation, imbalanced osteoimmune, and impaired vascular system in the diabetic microenvironment will result in a high rate of impaired, delayed, or even nonhealing events of bone tissue. Stimuli-responsive biomaterials that can respond to endogenous biochemical signals have emerged as effective therapeutic systems to treat diabetic bone defects via the combination of microenvironmental regulation and enhanced osteogenic capacity. Following the natural bone healing processes, coupling of angiogenesis and osteogenesis by advanced bioresponsive drug delivery systems has proved to be of significant approach for promoting bone repair in DM. In this Review, we have systematically summarized the mechanisms and therapeutic strategies of DM-induced impaired bone healing, outlined the bioresponsive design for drug delivery systems, and highlighted the vascularization strategies for promoting bone regeneration. Accordingly, we then overview the recent advances in developing bioresponsive drug delivery systems to facilitate diabetic vascularized bone regeneration by remodeling the microenvironment and modulating multiple regenerative cues. Furthermore, we discuss the development of adaptable drug delivery systems with unique features for guiding DM-associated bone regeneration in the future.
Collapse
Affiliation(s)
- Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Andrij Pich
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
35
|
Yuan L, Liu J, Xiao S, Wei J, Liu H, Li Y, Zuo Y, Li Y, Wang J, Li J. EGCG-Modified Bioactive Core-Shell Fibers Modulate Oxidative Stress to Synergistically Promote Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:543-555. [PMID: 39743979 DOI: 10.1021/acsbiomaterials.4c01906] [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: 01/04/2025]
Abstract
Oxidative stress induced by reactive oxygen species (ROS) can adversely affect tissue repair, whereas endowing biomaterials with antioxidant activity can improve the in vivo microenvironment, thereby promoting angiogenesis and osteogenesis. Accordingly, this study utilized epigallocatechin-3-gallate (EGCG), a material known for its reducing properties, oxidative self-polymerization capability, and strong binding characteristics, to modify a bioactive core-shell fibrous membrane (10RP-PG). Compared to the 10RP-PG fibrous membrane, the EGCG-modified fibrous membrane (E/10RP-PG) exhibited superior hydrophilicity, excellent cell adhesion, and compatibility. Moreover, the EGCG-modified fibrous membrane can effectively scavenge free radicals, ameliorate the local microenvironment, and foster angiogenesis (enhancing the expression of angiogenic genes in human umbilical vein endothelial cells (HUVECs) by 1.58 times and promoting vascular generation area upon subcutaneous implantation by 4.47 times). The enhancement of angiogenic activity of the E/10RP-PG fibrous membrane further promoted cartilage degeneration and absorption, as well as new bone formation, thus facilitating the repair of bone defects. This study provides a new strategy for promoting bone defect repair through the surface modification of biomaterials with an antioxidant agent, and the fabricated E/10RP-PG fibrous membranes show promise for guiding vascularized bone regeneration.
Collapse
Affiliation(s)
- Li Yuan
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Jiangshan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Shiqi Xiao
- Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu 610081, PR China
| | - Jiawei Wei
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Huan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yongzhi Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, PR China
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
36
|
Xu YR, Tang DY, Xiao ZP, Huang ZT, Zhang HR, Tang ZW, He F. Effects of polylactic acid scaffolds with various orientations and diameters on osteogenesis and angiogenesis. Front Bioeng Biotechnol 2025; 12:1495810. [PMID: 39834633 PMCID: PMC11743647 DOI: 10.3389/fbioe.2024.1495810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Researchers in the field of regenerative medicine have consistently focused on the biomimetic design of engineered bone materials on the basis of the microstructure of natural bone tissue. Additionally, the effects of the micromorphological characteristics of these materials on angiogenesis have garnered increasing attention. In vitro, the orientation and diameter of scaffold materials can exert different effects on osteogenesis and vascularisation. However, more comprehensive investigations, including in vivo studies, are required to confirm the results observed in vitro. Accordingly, in the present study, fibre scaffolds with various orientations and diameters were prepared by electrospinning with polylactic acid. The effects of the micromorphological characteristics of these scaffolds with different orientations and diameters on osteogenesis and vascularisation were systematically studied via in vivo experiments. The scaffolds with aligned micromorphological features positively affected osteogenesis and vascularisation, which indicated that such characteristics could be considered crucial factors when designing materials for bone repair.
Collapse
Affiliation(s)
- Yun Rong Xu
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Dai Yuan Tang
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Zhen Ping Xiao
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Zai Tian Huang
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Heng Rui Zhang
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Zi Wen Tang
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fei He
- Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| |
Collapse
|
37
|
Park H, Trupiano SP, Medarev SL, Ghosh P, Caldwell JT, Yarrow JF, Muller-Delp JM. Aerobic exercise training-induced bone and vascular adaptations in mice lacking adiponectin. Bone 2025; 190:117272. [PMID: 39369833 PMCID: PMC11795456 DOI: 10.1016/j.bone.2024.117272] [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: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Adiponectin regulates lipid and glucose metabolism, and insulin sensitivity in various target organs; however, the effects of adiponectin on bone health remain controversial. Exercise training can enhance bone density, bone microarchitecture, and blood flow. This study aimed to elucidate the role of adiponectin in adaptations of bone microarchitecture and bone vasculature in response to aerobic exercise training. Adult male C57BL/6 wild-type (WT) and homozygous adiponectin knockout (AdipoKO) mice were either treadmill exercise trained or remained sedentary for 8-10 weeks. The trabecular structures of the distal femoral metaphysis, a weight-bearing bone, and the mandible, a non-weight-bearing bone, were examined using microcomputed tomography. The femoral principal nutrient arteries were isolated to assess vasoreactivity (vasodilation and vasoconstriction) and structural remodeling. At the femoral metaphysis, impaired trabecular bone structures, including reduced connectivity density and increased trabecular spacing, were observed in AdipoKO mice compared to WT mice. In addition, nitric oxide-mediated, endothelium-dependent vasodilation was substantially reduced, and wall-to-lumen ratio was significantly increased in the femoral principal nutrient artery of AdipoKO mice. Interestingly, although exercise training-induced enhancements in trabecular connectivity density were observed at the femoral metaphysis of both WT and AdipoKO, increased vasoconstrictor responses were only observed in the femoral principal nutrient artery of WT mice, not in the AdipoKO mice. In mandibular trabecular bone, exercise training increased trabecular bone volume fraction (BV/TV, %) and intersection surface in the mandible of both WT and AdipoKO mice. These findings indicate that adiponectin is crucial for maintaining normal bone microarchitecture and vasculature. Although the absence of adiponectin compromises bone vascular adaptation to exercise training in mice, some exercise training-induced alterations in bone microarchitecture occurred in the absence of adiponectin, suggesting contribution of compensatory mechanisms during exercise training.
Collapse
Affiliation(s)
- Hyerim Park
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Samuel P Trupiano
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Steven L Medarev
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Payal Ghosh
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Jacob T Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA; Department of Exercise and Sport Science, University of Wisconsin-La Crosse, La Crosse, WI, USA
| | - Joshua F Yarrow
- Eastern Colorado Geriatrics Research, Education, and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO, USA
| | - Judy M Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
38
|
Ayoub M, Susin SA, Bauvois B. Tumor Cell Survival Factors and Angiogenesis in Chronic Lymphocytic Leukemia: How Hot Is the Link? Cancers (Basel) 2024; 17:72. [PMID: 39796700 PMCID: PMC11719013 DOI: 10.3390/cancers17010072] [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: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic CD5+/CD19+ B lymphocytes in the blood. These cells migrate to and proliferate in the bone marrow and lymphoid tissues. Despite the development of new therapies for CLL, drug resistance and disease relapse still occur; novel treatment approaches are therefore still needed. Inhibition of the angiogenesis involved in the progression of CLL might be a relevant therapeutic strategy. The literature data indicate that vascular endothelial growth factor, angiopoietin-2, and matrix metalloproteinase-9 are pro-angiogenic factors in CLL. A number of other CLL factors might have pro-angiogenic activity: fibroblast growth factor-2, certain chemokines (such as CXCL-12 and CXCL-2), tumor necrosis factor-α, insulin-like growth factor-1, neutrophil gelatinase-associated lipocalin, and progranulin. All these molecules contribute to the survival, proliferation, and migration of CLL cells. Here, we review the literature on these factors' respective expression profiles and roles in CLL. We also summarize the main results of preclinical and clinical trials of novel agents targeting most of these molecules in a CLL setting. Through the eradication of leukemic cells and the inhibition of angiogenesis, these therapeutic approaches might alter the course of CLL.
Collapse
Affiliation(s)
| | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS 1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France; (M.A.); (S.A.S.)
| |
Collapse
|
39
|
Lee JE, Hwa S, Lee HR, Kim JH, Lee HJ, Park JB. Impact of Vascular Endothelial Growth Factor on the Shape, Survival, and Osteogenic Transformation of Gingiva-Derived Stem Cell Spheroids. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2108. [PMID: 39768988 PMCID: PMC11677937 DOI: 10.3390/medicina60122108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/07/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Vascular endothelial growth factor (VEGF) is a protein which stimulates the formation of new blood vessels, playing a crucial role in processes such as wound healing and tumor growth. Methods: This study investigated the effects of VEGF on cell viability and osteogenic differentiation in mesenchymal stem cell (MSC) spheroids. Stem cell spheroids were fabricated using concave microwells and cultured with VEGF at concentrations of 0, 0.01, 0.1, 1, and 10 ng/mL. Morphological assessments were conducted on days 1, 3, 5, and 7, while cell viability was evaluated using the LIVE/DEAD assay and Cell Counting Kit-8. Alkaline phosphatase activity (ALP) and calcium deposition were measured to assess osteogenic differentiation, and qPCR was used to analyze osteogenic marker expression. Results: The spheroids maintained their shape across all VEGF concentrations, with the largest diameter being at 0.01 ng/mL on day 1, which decreased over time. Cell viability was highest at 0.01 ng/mL VEGF, while calcium deposition peaked at 0.1 ng/mL. Osteogenic markers, including RUNX2, osteocalcin, and COL1A1, showed significant upregulation at 1 ng/mL VEGF. Conclusions: These results suggest that VEGF enhances early osteogenic differentiation in MSC spheroids, indicating its potential for bone repair and tissue regeneration. VEGF could be applied in clinical settings for bone healing, fracture repair, and regenerative dentistry treatments.
Collapse
Affiliation(s)
- Ji-Eun Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
- Department of Periodontics, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Somyeong Hwa
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hee-Ra Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ju-Hwan Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
| | - Hyun-Jin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-E.L.); (S.H.); (H.-R.L.); (J.-H.K.); (H.-J.L.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
40
|
Zhong C, Tang Z, Yu X, Wang L, Ren C, Qin L, Zhou P. Advances in the Construction and Application of Bone-on-a-Chip Based on Microfluidic Technologies. J Biomed Mater Res B Appl Biomater 2024; 112:e35502. [PMID: 39555794 DOI: 10.1002/jbm.b.35502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bone-on-a-chip (BOC) models that based on microfluidic technology have widely applied to understand bone physiology and the pathogenesis of related diseases. In this review, we provide an overview of bone biology and related diseases, explain the advantages and applications of microfluidic technology in the construction of BOC models, and summarize their progress in physiology, pathology, and drug development. Finally, we discussed the problems to be solved and the future directions of microfluidic technology and BOC platforms, so as to provide a reference for researchers to design better BOC models.
Collapse
Affiliation(s)
- Chang Zhong
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Zihui Tang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Xin Yu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Lu Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Chenyuan Ren
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Gansu Health Vocational College, Lanzhou, China
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
41
|
Asayag K, Peled E, Assalia M, Crispel Y, Yanovich C, Cohen H, Keren-Politansky A, Nadir Y. Steroids and Malignancy Increase Local Heparanase and Decrease Markers of Osteoblast Activity in Bone Tissue Microcirculation. Biomolecules 2024; 14:1506. [PMID: 39766213 PMCID: PMC11673960 DOI: 10.3390/biom14121506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/18/2024] [Accepted: 11/23/2024] [Indexed: 01/30/2025] Open
Abstract
Bone metastasis and steroids are known to activate the coagulation system and induce osteoporosis, pathological bone fractures, and bone pain. Heparanase is a protein known to enhance the hemostatic system and to promote angiogenesis, metastasis, and inflammation. The objective of the present study was to evaluate the effects of steroids and malignancy on the coagulation factors and osteoblast activity in the bone tissue. The effects of dexacort and malignant medium were evaluated in osteoblasts derived from human bone marrow mesenchymal stem cells and human umbilical vein endothelial cells (HUVECs). The bones of mice treated with dexacort for 1 month were studied. Bone biopsies of ten patients with bone metastasis, ten with steroid-induced avascular necrosis (AVN), and ten with osteoarthritis were compared to ten controls. We found that dexacort and malignant medium significantly increased the heparanase levels in osteoblasts and HUVECs and decreased the levels of alkaline phosphatase (ALKP). Peptide 16AC, derived from heparanase, which interacts with tissue factor (TF), further increased the effect, while peptide 6, which inhibits interactions between heparanase and TF, reversed the effect in these cells. The bone microcirculation of mice treated with dexacort exhibited significantly higher levels of heparanase, TF, TF pathway inhibitor (TFPI), TFPI-2, thrombin, and syndecan-1, but reduced levels of osteocalcin and ALKP. The pathological human bone biopsies' microcirculation exhibited significantly dilated blood vessels and higher levels of heparanase, TF, TFPI, TFPI-2, and fibrin. In summary, steroids and malignancy increased the activation of the coagulation system in the bone microcirculation and reduced the osteoblast activity. Heparanase inhibitors should be further investigated to attenuate bone fractures and pain.
Collapse
Affiliation(s)
- Keren Asayag
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Eli Peled
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
- Orthopedic Division, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Mai Assalia
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Yonatan Crispel
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Chen Yanovich
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Haim Cohen
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Anat Keren-Politansky
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| | - Yona Nadir
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, Haifa 3109601, Israel; (K.A.); (M.A.); (Y.C.); (C.Y.); (H.C.); (A.K.-P.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Israel Institute of Technology—Technion, Haifa 3109601, Israel;
| |
Collapse
|
42
|
Wei S, Hu Q, Dong J, Sun Y, Bai J, Shan H, Gao X, Sheng L, Dai J, Jiang F, Dai X, Gu X, Zhou X. Mechanically enhanced biodegradable scaffold based on SF microfibers for repairing bone defects in the distal femur of rats. Int J Biol Macromol 2024; 282:137372. [PMID: 39521213 DOI: 10.1016/j.ijbiomac.2024.137372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Silk-based biodegradable materials play an important role in tissue engineering, especially in the field of bone regeneration. However, while optimizing mechanical properties and bone regeneration characteristics, modified silk fibroin (SF)-based materials also increase the complexity of scaffold systems, which is not conducive to clinical translation. In this study, we first added synthetic biomimetic mineralized collagen (MC) particles to SF-based materials to improve the bone regeneration properties of the scaffolds and simultaneously regulated the degradation rate of the scaffolds to match the bone regeneration rate. Second, SF microfibers were prepared by hydrolysis with alkaline heating and added to SFMC scaffolds with excellent osteogenic stimulation ability to prepare SF microfiber (mf)-modified SFMC-mf scaffolds with excellent mechanical properties, whose compression modulus increased from 4.58±0.23 MPa to 14.63±0.88 MPa. Finally, the SFMC-mf scaffold was implanted into the weight-bearing bone defect area of the distal femur of rats, and the results showed that the SFMC-mf scaffold significantly promoted functional recovery of the affected limb and increased the amount of new bone in the defect area compared with those in the SFC-mf group and the blank control group. In addition, the RNA-seq results suggested that the genes with upregulated expression in the SFMC-mf scaffold group were mainly enriched in vascular regeneration. In conclusion, this SF microfiber modification method effectively improved the mechanical properties of SFMC scaffolds without moving the SF scaffold system in the direction of compositional complexity, providing new insights for the subsequent development of more effective bionic repair materials for bone defects and assisting in their clinical translation.
Collapse
Affiliation(s)
- Shuai Wei
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Qian Hu
- Health Management Center, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jin Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Yu Sun
- New Material Technology, Soochow Xianjue, Suzhou, Jiangsu 215000, China
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Huajian Shan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jun Dai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Fengxian Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
43
|
Sengokmen-Ozsoz N, Aleemardani M, Palanca M, Hann A, Reilly GC, Dall'Ara E, Claeyssens F. Fabrication of hierarchically porous trabecular bone replicas via 3D printing with high internal phase emulsions (HIPEs). Biofabrication 2024; 17:015012. [PMID: 39454611 DOI: 10.1088/1758-5090/ad8b70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
Combining emulsion templating with additive manufacturing enables the production of inherently porous scaffolds with multiscale porosity. This approach incorporates interconnected porous materials, providing a structure that supports cell ingrowth. However, 3D printing hierarchical porous structures that combine semi-micropores and micropores remains a challenging task. Previous studies have demonstrated that using a carefully adjusted combination of light absorbers and photoinitiators in the resin can produce open surface porosity, sponge-like internal structures, and a printing resolution of about 150µm. In this study, we explored how varying concentrations of tartrazine (0, 0.02, 0.04, and 0.08 wt%) as a light absorber affect the porous structure of acrylate-based polymerized medium internal phase emulsions fabricated via vat photopolymerization. Given the importance of a porous and interconnected structure for tissue engineering and regenerative medicine, we tested cell behavior on these 3D-printed disk samples using MG-63 cells, examining metabolic activity, adhesion, and morphology. The 0.08 wt% tartrazine-containing 3D-printed sample (008 T) demonstrated the best cell proliferation and adhesion. To show that this high internal phase emulsion (HIPE) resin can be used to create complex structures for biomedical applications, we 3D-printed trabecular bone structures based on microCT imaging. These structures were further evaluated for cell behavior and migration, followed by microCT analysis after 60 days of cell culture. This research demonstrates that HIPEs can be used as a resin to print trabecular bone mimics using additive manufacturing, which could be further developed for lab-on-a-chip models of healthy and diseased bone.
Collapse
Affiliation(s)
- Nihan Sengokmen-Ozsoz
- Kroto Research Institute, Department of Materials Science and Engineering, The University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Department of Materials Science and Engineering, Gebze Technical University, Gebze, Kocaeli 41400, Turkey
| | - Mina Aleemardani
- Kroto Research Institute, Department of Materials Science and Engineering, The University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Department of Translational Health Science, Bristol Medical School, University of Bristol, Bristol BS1 3NY, United Kingdom
| | - Marco Palanca
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Industrial Engineering, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Alice Hann
- Department of Materials Science and Engineering, Pam Liversidge Building, Mappin Street, Sheffield, United Kingdom
| | - Gwendolen C Reilly
- Department of Materials Science and Engineering, Pam Liversidge Building, Mappin Street, Sheffield, United Kingdom
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| | - Frederik Claeyssens
- Kroto Research Institute, Department of Materials Science and Engineering, The University of Sheffield, Sheffield S3 7HQ, United Kingdom
| |
Collapse
|
44
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024; 13:e2401307. [PMID: 39175382 PMCID: PMC11582516 DOI: 10.1002/adhm.202401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Janis Locs
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| |
Collapse
|
45
|
Hosseini SF, Galefi A, Hosseini S, Shaabani A, Farrokhi N, Jahanfar M, Nourany M, Homaeigohar S, Alipour A, Shahsavarani H. Magnesium oxide nanoparticle reinforced pumpkin-derived nanostructured cellulose scaffold for enhanced bone regeneration. Int J Biol Macromol 2024; 281:136303. [PMID: 39370065 DOI: 10.1016/j.ijbiomac.2024.136303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Considering global surge in bone fracture prevalence, limitation in use of traditional healing approaches like bone grafts highlights the need for innovative regenerative strategies. Here, a novel green fabrication approach has reported for reinforcement of physicochemical performances of sustainable bioinspired extracellular matrix (ECM) based on decellularized pumpkin tissue coated with Magnesium oxide nanoparticles (hereafter called DM-Pumpkin) for enhanced bone regeneration. Compared to uncoated scaffold, DM-Pumpkin exhibited significantly improved surface roughness, mechanical stiffness, porosity, hydrophilicity, swelling, and biodegradation rate. Obtained nanoporous structure provides an ideal three-dimensional microenvironment for the attachment, migration and osteo-induction in human adipose-derived mesenchymal stem cells (h- AdMSCs). Calcium deposition and mineralization, alkaline phosphatase activity, and SEM imaging of the cells as well as increased expression of bone-related genes after 21 days incubation confirmed capability of DM-Pumpkin in mimicking the biological properties of bone tissue. The presence of MgONPs had a silencing effect on inflammatory factors and improved wound closure, verified by in vivo studies. Increased expression of collagen type I and osteocalcin in the h- AdMSCs cultured on DM-Pumpkin compared to control further corroborated gained results. Altogether, boosting physicochemical and biological properties of DM-Pumpkin due to surface modification is a promising approach for guided bone regeneration.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Hosseini
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran; Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Atena Galefi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Alireza Shaabani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, GC, 1983969411 Tehran, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Mehdi Jahanfar
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Mohammad Nourany
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran; Faculty of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Atefeh Alipour
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran; Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran.
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran; Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran 1316943551, Iran; Iranian Biological Resource Center, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran.
| |
Collapse
|
46
|
Gao N, Tian M, Sun Y, Wang D, Sun G. Application of bone perforation in the surgery of medication-related osteonecrosis of the jaw in stage Ⅱ. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2024; 42:629-635. [PMID: 39304506 PMCID: PMC11493859 DOI: 10.7518/hxkq.2024.2024133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/30/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES This study aimed to explore the effect of surgery combined with bone perforation for treating stage Ⅱ medication-related osteonecrosis of the jaw (MRONJ). METHODS A total of 21 patients with stage Ⅱ mandibular MRONJ who underwent surgical treatment from June 2020 to June 2023 were included in this study. Retrospective analysis was conducted on their clinical data, including gender, age, primary disease, drug name and administration method, pre-surgery drug cessation, and prognosis. The cohort comprised 14 males and 7 females, with an average age at onset of 68.33±10.74 years. According to the guidelines of the American Association of Oral and Maxillofacial Surgeons, the included patients had stage Ⅱ mandibular MRONJ. The treatment approach consisted of partial mandibulectomy combined with bone perforation techniques, ensuring tension-free suturing of soft tissues. Follow-up was performed regularly, and the curative effect was evaluated. The SF-12 health survey was used to assess the quality of life for all patients before and after surgery. RESULTS A total of 21 patients were followed up for 8-38 months after surgery, and the mucosal healing of 17 patients was good (80.95%). The postoperative quality of life score (83.62±5.90) was significantly higher than that before operation (63.67±4.70, P<0.05). CONCLUSIONS Surgery combined with bone perforation te-chnique is an effective treatment method with high success rate in refractory stage Ⅱ MRONT patients.
Collapse
Affiliation(s)
- Na Gao
- Dept. of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Mei Tian
- Dept. of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Yawei Sun
- Dept. of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Danni Wang
- Dept. of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Guowen Sun
- Dept. of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing 210008, China
| |
Collapse
|
47
|
Yang S, Wu H, Peng C, He J, Pu Z, Lin Z, Wang J, Hu Y, Su Q, Zhou B, Yong X, Lan H, Hu N, Hu X. From the microspheres to scaffolds: advances in polymer microsphere scaffolds for bone regeneration applications. BIOMATERIALS TRANSLATIONAL 2024; 5:274-299. [PMID: 39734699 PMCID: PMC11681185 DOI: 10.12336/biomatertransl.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
The treatment and repair of bone tissue damage and loss due to infection, tumours, and trauma are major challenges in clinical practice. Artificial bone scaffolds offer a safer, simpler, and more feasible alternative to bone transplantation, serving to fill bone defects and promote bone tissue regeneration. Ideally, these scaffolds should possess osteoconductive, osteoinductive, and osseointegrative properties. However, the current first-generation implants, represented by titanium alloys, have shown poor bone-implant integration performance and cannot meet the requirements for bone tissue repair. This has led to increased research on second and third generation artificial bone scaffolds, which focus on loading bioactive molecules and cells. Polymer microspheres, known for their high specific surface areas at the micro- and nanoscale, exhibit excellent cell and drug delivery behaviours. Additionally, with their unique rigid structure, microsphere scaffolds can be constructed using methods such as thermal sintering, injection, and microsphere encapsulation. These scaffolds not only ensure the excellent cell drug loading performance of microspheres but also exhibit spatial modulation behaviour, aiding in bone repair within a three-dimensional network structure. This article provides a summary and discussion of the use of polymer microsphere scaffolds for bone repair, focusing on the mechanisms of bone tissue repair and the current status of clinical bone grafts, aimed at advancing research in bone repair.
Collapse
Affiliation(s)
- Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Haoming Wu
- School of Preclinical Medicine of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Chao Peng
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Zhengguang Pu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jun Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yingkun Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Bingnan Zhou
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
| | - Hai Lan
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
48
|
Huang J, Liao C, Yang J, Zhang L. The role of vascular and lymphatic networks in bone and joint homeostasis and pathology. Front Endocrinol (Lausanne) 2024; 15:1465816. [PMID: 39324127 PMCID: PMC11422228 DOI: 10.3389/fendo.2024.1465816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
The vascular and lymphatic systems are integral to maintaining skeletal homeostasis and responding to pathological conditions in bone and joint tissues. This review explores the interplay between blood vessels and lymphatic vessels in bones and joints, focusing on their roles in homeostasis, regeneration, and disease progression. Type H blood vessels, characterized by high expression of CD31 and endomucin, are crucial for coupling angiogenesis with osteogenesis, thus supporting bone homeostasis and repair. These vessels facilitate nutrient delivery and waste removal, and their dysfunction can lead to conditions such as ischemia and arthritis. Recent discoveries have highlighted the presence and significance of lymphatic vessels within bone tissue, challenging the traditional view that bones are devoid of lymphatics. Lymphatic vessels contribute to interstitial fluid regulation, immune cell trafficking, and tissue repair through lymphangiocrine signaling. The pathological alterations in these networks are closely linked to inflammatory joint diseases, emphasizing the need for further research into their co-regulatory mechanisms. This comprehensive review summarizes the current understanding of the structural and functional aspects of vascular and lymphatic networks in bone and joint tissues, their roles in homeostasis, and the implications of their dysfunction in disease. By elucidating the dynamic interactions between these systems, we aim to enhance the understanding of their contributions to skeletal health and disease, potentially informing the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingxiong Huang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| | - Chengcheng Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Guizhou, Zunyi, China
| | - Jian Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Zhang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
49
|
Barzilay JI, Buzkova P, Bielinski SJ, Cotch MF, Kestenbaum B, Austin TR, Carbone L, Mukamal KJ, Budoff MJ. The association of microvascular disease and endothelial dysfunction with vertebral trabecular bone mineral density : The MESA study. Osteoporos Int 2024; 35:1595-1604. [PMID: 38913124 DOI: 10.1007/s00198-024-07152-y] [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: 02/26/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
Retinopathy and albuminuria are associated with hip fracture risk. We investigated whether these disorders and endothelial dysfunction (which underlies microvascular diseases) were associated with low trabecular bone density. No significant associations were found, suggesting that microvascular diseases are not related to fracture risk through low trabecular bone density. PURPOSE Microvascular diseases of the eye, kidney, and brain are associated with endothelial dysfunction and increased hip fracture risk. To explore the basis for higher hip fracture risk, we comprehensively examined whether markers of microvascular disease and/or endothelial dysfunction are related to trabecular bone mineral density (BMD), a proximate risk factor for osteoporotic fractures. METHODS Among 6814 participants in the Multi-Ethnic Study of Atherosclerosis study (MESA), we derived thoracic vertebral trabecular BMD from computed tomography of the chest and measured urine albumin to creatinine ratios (UACR), retinal arteriolar and venular widths, flow mediated dilation (FMD) of the brachial artery after 5 min of ischemia; and levels of five soluble endothelial adhesion markers (ICAM-1, VCAM-1, L-selectin, P-selectin, and E-selectin). Linear regression models were used to examine the association of trabecular BMD with markers of microvascular disease and with markers of endothelial dysfunction. RESULTS We observed no significant associations of UACR, retinal arteriolar or venular widths, or FMD with BMD. We also observed no statistically significant association of spine trabecular BMD with levels of endothelial adhesion markers. Men and women had largely similar results. CONCLUSION We conclude that there is little evidence to connect thoracic spine trabecular BMD to microvascular disorders or to endothelial dysfunction among multi-ethnic middle-aged and older adults. Other factors beyond trabecular BMD (e.g., bone quality or predisposition to falling) may be responsible for the associations of microvascular disease with osteoporotic fractures.
Collapse
Affiliation(s)
- Joshua I Barzilay
- Division of Endocrinology, Kaiser Permanente of Georgia, 3650 Steve Reynolds Blvd, Duluth, GA, 30096, USA.
- Division of Endocrinology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Petra Buzkova
- Division of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Mary Frances Cotch
- Office of Vision Health and Population Sciences, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bryan Kestenbaum
- Division of Nephrology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Department of Epidemiology, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Laura Carbone
- Division of Rheumatology, Medical College of Georgia, Augusta, GA, USA
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Brookline, MA, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute at Harbor-UCLA, Torrance, CA, USA
| |
Collapse
|
50
|
Zang C, Che M, Xian H, Xiao X, Li T, Chen Y, Liu Y, Cong R. 3D-printed silicate porous bioceramics promoted the polarization of M2-macrophages that enhanced the angiogenesis in bone regeneration. J Biomed Mater Res B Appl Biomater 2024; 112:e35469. [PMID: 39205328 DOI: 10.1002/jbm.b.35469] [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: 11/27/2023] [Revised: 07/16/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
The failure of bone regeneration has been considered as a serious problem that troubling patients for decades, most of which was resulted by the poor angiogenesis and chronic inflammation after surgery. Among multiple materials applied in the repair of bone defect, silicate bioceramics attracted researchers because of its excellent bioactivity. The purpose of this study was to detect the effect of specific bioactive glass ceramic (AP40, based on crystalline phases of apatite and wollastonite) on angiogenesis and the subsequent bone growth through the modulation of macrophages. Two groups were included in this study: control group (macrophages without any stimulation, denominated as Control) and AP40 group (macrophages incubated on AP40). This study investigated the effect of AP40 on macrophages polarization (RAW264.7) and angiogenesis in vitro and in vivo. Additionally, the changes of angiogenic ability regulated by macrophages were explored. AP40 showed excellent angiogenesis potential and the expression of CD31 was promoted through the modulation of macrophages toward M2 subtype. Additionally, the macrophages incubated on AP40 synthesized more PDGF-BB comparing to macrophages without any stimulation, which contributed to the improved angiogenetic ability of human umbilical vein endothelial cells (HUVECs). Results of in vivo studies indicated increased bone ingrowth along the implants, which indicated the potential of bioceramics for bone defect repair clinically.
Collapse
Affiliation(s)
- Chengwu Zang
- Department of Orthopedics, Xijing Hospital, the Air Force Medical University, Xi'an, China
| | - Min Che
- Affiliated Central Hospital of Shenyang Medical College, Shenyang, China
| | - Hang Xian
- Department of Orthopedics, Xijing Hospital, the Air Force Medical University, Xi'an, China
| | - Xin Xiao
- Department of Orthopedics, Xijing Hospital, the Air Force Medical University, Xi'an, China
| | - Tengfei Li
- The North West Institute of Nuclear Technology, Xi'an, China
| | - Yongxiang Chen
- Department of Orthopedics, Xijing Hospital, the Air Force Medical University, Xi'an, China
| | | | - Rui Cong
- Department of Orthopedics, Xijing Hospital, the Air Force Medical University, Xi'an, China
| |
Collapse
|