1
|
Silveira A, Davydok A, Krywka C, Scheel M, Weitkamp T, Fleck C, Shahar R, Zaslansky P. Nanocrystal Compressive Residual Stresses: A Strategy to Strengthen the Bony Spines of Osteocytic and Anosteocytic Fish. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410617. [PMID: 40213975 PMCID: PMC12120745 DOI: 10.1002/advs.202410617] [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] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/07/2025] [Indexed: 06/01/2025]
Abstract
Bone is a living tissue in which communicating cells, osteocytes, are assumed to be vital for tissue turnover and adaptation. Interestingly however, most advanced teleost fish do not possess osteocyte-mediated porosity, prompting intriguing questions about alternative material-strategies for these bones to cope with damage. Using advanced imaging techniques, including phase-contrast enhanced (PCE) microtomography (µCT) and nanotomography (nanoCT), X-ray fluorescence (XRF), and diffraction (XRD) tomography, the micro- and nano-architectures of osteocytic zebrafish are compared with anosteocytic medaka fishbone. PCE µCT and Zernike phase-contrast nanoCT showed a lack of porosity in medaka bone and 0.75 - 2.26% osteocytic porosity in zebrafish. Both fish species have similar mineralized collagen fibril arrangements containing calcium (Ca) and traces of strontium (Sr) with increased zinc (Zn) localized on the outer bone regions. Medaka bones exhibit reduced apatite nanocrystal lattice spacings on the outer surfaces. Indeed we find higher compressive residual strains (-0.100 ± 0.02) compared to zebrafish (-0.071 ± 0.03). We propose that medaka bone evolved to replace the mechanosensitive osteocytic network by entrapping protective residual strains between collagen nanofibers and mineral crystals. These strains may enhance fracture toughness while making this nanocomposite well-suited for sustaining repeated loading cycles, thus reducing the metabolic costs associated with housing a large network of cells.
Collapse
Affiliation(s)
- Andreia Silveira
- Department for RestorativePreventive and Pediatric DentistryCharité‐Universitaetsmedizin14197BerlinGermany
| | - Anton Davydok
- Institute of Materials PhysicsHelmholtz‐Zentrum Hereon21502GeesthachtGermany
| | - Christina Krywka
- Institute of Materials PhysicsHelmholtz‐Zentrum Hereon21502GeesthachtGermany
| | | | | | - Claudia Fleck
- Fachgebiet Werkstofftechnik/Chair of Materials Science & EngineeringInstitute of Materials Science and TechnologyFaculty III ‐ Process SciencesTechnische Universität Berlin10623BerlinGermany
| | - Ron Shahar
- Koret School of Veterinary MedicineThe Robert H. Smith Faculty of AgricultureFood and Environmental SciencesHebrew University of JerusalemRehovot76100Israel
| | - Paul Zaslansky
- Department for RestorativePreventive and Pediatric DentistryCharité‐Universitaetsmedizin14197BerlinGermany
| |
Collapse
|
2
|
Zhang L, Zhou Z, Liu Z, Shi Y, Mao Y, Fu B, Wang Z. Alendronate and polyelectrolyte synergically induce biomimetic mineralization of collagen and demineralized dentin. Int J Biol Macromol 2025; 308:142402. [PMID: 40154709 DOI: 10.1016/j.ijbiomac.2025.142402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Alendronate (ALN), a potent bisphosphonate, plays a significant role in both bone formation and osteoporosis therapy. However, its potential to promote biomimetic mineralization of type I collagen fibrils and demineralized dentin remains unclear. This study reveals that ALN-pretreated collagen fibrils form ester bonds between the hydroxyl groups of ALN and the carboxyl groups of collagen, accompanied by a reduction in collagen surface potential. Additionally, an adsorption equilibrium of ALN to collagen was achieved at 25 mM ALN. ALN pretreatment facilitates intrafibrillar mineralization of type I collagen fibrils in a dose-dependent manner, as well as mineralization of demineralized dentin in synergy with polyelectrolyte-stabilized Amorphous Calcium Phosphate (ACP) nanoparticles. This process effectively restores the mechanical properties of demineralized dentin to levels comparable to natural dentin. Thus, ALN holds potential for localized applications aimed at promoting remineralization of demineralized dentin.
Collapse
Affiliation(s)
- Leiqing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zihuai Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwen Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Ying Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Yingjie Mao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Baiping Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhe Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, China; Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, China; Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
3
|
Kollert MR, Krämer M, Brisson NM, Schemenz V, Tsitsilonis S, Qazi TH, Fratzl P, Vogel V, Reichenbach JR, Duda GN. Water and ions binding to extracellular matrix drives stress relaxation, aiding MRI detection of swelling-associated pathology. Nat Biomed Eng 2025; 9:772-786. [PMID: 40234703 PMCID: PMC12092267 DOI: 10.1038/s41551-025-01369-w] [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: 12/08/2022] [Accepted: 02/10/2025] [Indexed: 04/17/2025]
Abstract
Swelling-associated changes in extracellular matrix (ECM) occur in many pathological conditions involving inflammation or oedema. Here we show that alterations in the proportion of loosely bound water in ECM correlate with changes in ECM elasticity and stress relaxation, owing to the strength of water binding to ECM being primarily governed by osmolality and the electrostatic properties of proteoglycans. By using mechanical testing and small-angle X-ray scattering, as well as magnetic resonance imaging (MRI) to detect changes in loosely bound water, we observed that enhanced water binding manifests as greater resistance to compression (mechanical or osmotic), resulting from increased electrostatic repulsion between negatively charged proteoglycans rather than axial contraction in collagen fibrils. This indicates that electrostatic contributions of proteoglycans regulate elasticity and stress relaxation independently of hydration. Our ex vivo experiments in osmotically modulated tendon elucidate physical causes of MRI signal alterations, in agreement with pilot in vivo MRI of inflammatory Achilles tendinopathy. We suggest that the strength of water binding to ECM regulates cellular niches and can be exploited to enhance MRI-informed diagnostics of swelling-associated tissue pathology.
Collapse
Affiliation(s)
- Matthias R Kollert
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Martin Krämer
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Nicholas M Brisson
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Victoria Schemenz
- Department of Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Serafeim Tsitsilonis
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Georg N Duda
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Nie J, Wu X, Wei J, Li Y, Xie H, Wang W, Wang H, Ping H, Li B, Fu Z. Regulating Growth of Strontium Carbonate in Self-Assembled Chiral Chitin Matrices with Robust Mechanical Properties. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25849-25860. [PMID: 40249380 DOI: 10.1021/acsami.5c04469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The exoskeleton of arthropods exhibits a Bouligand structure, composed of a chitin matrix and calcium carbonate crystals, which confer exceptional mechanical properties. While many studies focus on the relationship between structure and performance, few investigate the mineral growth process within the Bouligand matrix. Here, chiral chitin films are prepared through evaporation-induced self-assembly of chitin nanowhiskers, and subsequently incubated in SrCO3 mineralizing solution. Initially, precursors deposit on the film surface and transform into mineralized points, which then radially expand outward along the surface and propagate inward until coalescing into a continuous mineral layer. The growth rate of these mineralized points is significantly enhanced by increasing the reaction temperature; at 60 °C, the growth rate is 13 times faster (650.4 μm2/min) compared to that at 25 °C (49.7 μm2/min). Finally, SrCO3/chitin composite bulks are fabricated by stacking and hot-pressing multiple mineralized chitin films, adhered using sodium alginate (SA) solution through spin coating. The resulting SrCO3/chitin@SA composites exhibit a bending strength of 64.2 MPa, representing a 27% increase over pure chitin bulk and a 105% increase over pure SrCO3 bulk. Our work provides a strategy for low-temperature fabrication of high-performance artificial composites.
Collapse
Affiliation(s)
- Jiawei Nie
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Xiaoqian Wu
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Jingjiang Wei
- Institute for Advanced Materials Deformation and Damage from Multi-Scale, Institute for Advanced Study, Chengdu University, Chengdu 610106, China
| | - Yidi Li
- State Key Laboratory of Precision Blasting, Jianghan University, Wuhan 430056, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Weimin Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Hao Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Hang Ping
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Bin Li
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
| | - Zhengyi Fu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
5
|
Zhai CY, Ma A, Wang W, Zhu T, Huanyu L, Lan W, Yu T, Lan J, Wang Z. In Vivo and In Vitro Study of a Multifunctional SF/nHAp Corrosion-Resistant Bio-Coating Prepared on MAO Magnesium Alloy via Ultrasonic Spraying. ACS Biomater Sci Eng 2025; 11:2290-2305. [PMID: 40066752 DOI: 10.1021/acsbiomaterials.4c02405] [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: 04/15/2025]
Abstract
Magnesium alloys are often used in bone repair surgeries due to their biodegradability and excellent elastic modulus, making them a promising alternative to traditional nondegradable implants like titanium alloys. However, their rapid degradation rate limits their use as implants in the body. To enhance the corrosion resistance and bioactivity of magnesium alloys, we applied an ultrasonic spray coating on microarc oxidized (MAO) AZ31 magnesium alloy, using a mixture of silk fibroin (SF) and nanohydroxyapatite (nHAp). This SF/nHAp composite embeds directly into the micropores on the MAO-treated surface without additional physical or chemical treatment, forming a stable interlocked coating structure. The effects of different spray parameters on coating adhesion and interface characteristics were investigated, leading to the development of a corrosion-resistant and highly biocompatible composite coating. Further biological evaluations were conducted through subcutaneous implantation, assessing the in vivo degradation of the samples and the surrounding tissue response from multiple perspectives. A novel concept of in vivo tissue-reactive coatings was proposed, suggesting that highly biocompatible coating materials, in the early stages postimplantation, enable surrounding fibrous tissues to closely adhere to the surface, thereby slowing material degradation. As a result, the highly bioactive MAO-SF/nHAp coating significantly enhances the corrosion resistance of magnesium alloys, reduces hydrogen evolution, promotes regeneration of surrounding tissues, and minimizes postimplant inflammation. This approach offers a new strategy to improve the biocompatibility and corrosion resistance of magnesium alloys in vivo, suggesting that the overall evaluation of biodegradable magnesium alloys should focus more on assessing in-body corrosion.
Collapse
Affiliation(s)
- Chuan Yao Zhai
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - AnQuan Ma
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - Wenhao Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - TianTian Zhu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - Liu Huanyu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - WeiPeng Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - TianJiao Yu
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - Jing Lan
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China
| | - ZhiFeng Wang
- Department of Pediatric Dentistry (Department of Preventive Dentistry), School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, No.44-1 Wenhua Road West, Jinan, Shandong 250012, China
| |
Collapse
|
6
|
Miao S, Guo J, Zhang Y, Liu P, Chen X, Han Q, Wang Y, Xuan K, Yang P, Tao F. Biomimetic Intrafibrillar Mineralization of Hierarchically Structured Amyloid-Like Fibrils. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416824. [PMID: 40195686 DOI: 10.1002/adma.202416824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/26/2025] [Indexed: 04/09/2025]
Abstract
Intrafibrillar mineralization is essential not only as a fundamental process in forming biological hard tissues but also as a foundation for developing advanced composite fibril-based materials for innovative applications. Traditionally, only natural collagen fibrils have been shown to enable intrafibrillar mineralization, presenting a challenge in designing ordered hierarchical fibrils from common protein aggregation that exhibit similar high intrafibrillar mineralization activity. In this study, a mechanically directed two-step transformation method is developed that converts phase-transitioned protein nanofilms into crystalline, hierarchical amyloid-like fibrils with multilayer structures, which effectively control the growth and lateral organization of hydroxyapatite within adaptive gaps. The resulting mineralized HSAF achieves a hardness of 0.616 ± 0.007 GPa and a modulus of 19.06 ± 3.54 GPa-properties closely resembling native hard tissues-and exhibits exceptionally high bioactivity in promoting both native bone tissue growth and further intrafibrillar mineralization, achieving 76.9% repair in a mice cranial defect model after 8 weeks and outperforming other regenerative materials. This remarkable performance, stemming from the unique structure and composition of the fibers, positions HSAF as a promising candidate for biomedical and engineering applications. These findings advance the understanding of biomineralization mechanisms and establish a foundation for developing high-bioactivity materials for hard tissue regeneration.
Collapse
Affiliation(s)
- Shuting Miao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Jing Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yuexin Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Peisheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaojie Chen
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Qian Han
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Yingbo Wang
- College of Chemical Engineering, Xinjiang Key Laboratory of Energy Storage and Photoelectroctalytic Materials, Xinjiang Normal University, 102 Xinyi Road, Urumqi, 830054, China
| | - Kun Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| | - Fei Tao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, Xi'an Key Laboratory of Polymeric Soft Matter, International Joint Research Center on Functional Fiber and Soft Smart Textile, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
7
|
Tang Q, Nie J, Fang W, Xie H, Wang W, Wang H, Ping H, Li B, Fu Z. Effects of Polyacrylic Acid with Different Molecular Weights on Stress Generation through Regulating the Growth of Calcium Carbonate within Collagen. ACS APPLIED BIO MATERIALS 2025; 8:2386-2396. [PMID: 40020212 DOI: 10.1021/acsabm.4c01878] [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: 03/18/2025]
Abstract
Mineralized collagen fibrils are the building blocks of bone, and the mineralization of collagen fibrils is generally regulated by noncollagenous proteins (NCPs). However, the functions of NCPs are difficult to investigate in vivo. Here, we use poly(acrylic acid) (PAA) with different molecular weights (5, 50, 450, and 4000 kDa) as analogs of NCPs and explore their effects on collagen mineralization in vitro. All the PAA molecules can promote the intrafibrillar mineralization of calcium carbonate (CaCO3) following these steps: the precursors infiltrate the gap zones of collagen, and transform into organized calcite nanocrystals within collagen. An increase in molecular weight significantly accelerates the mineralization rate of collagen films, approximately 0.67 μm min-1 at 4000 kDa, four times that of 5 kDa (0.16 μm min-1). However, the generation of contractile stress via intrafibrillar mineralization in tendons exhibits a contrary tendency. It reaches 24.2 MPa at 5 kDa, much higher than that of 4000 kDa (8.3 MPa), due to rapid mineralization causing severe extrafibrillar precipitation around the tendon. The controllable mineralization of collagen matrices may inspire the development of bone repair and regeneration in the future.
Collapse
Affiliation(s)
- Qianshen Tang
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Jiawei Nie
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Weijian Fang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Weimin Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Hao Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Hang Ping
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| | - Bin Li
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, China
| | - Zhengyi Fu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan 430070, China
| |
Collapse
|
8
|
Hu J, Zheng K, Sherlock BE, Zhong J, Mansfield J, Green E, Toms AD, Winlove CP, Chen J. Zonal Characteristics of Collagen Ultrastructure and Responses to Mechanical Loading in Articular Cartilage. Acta Biomater 2025; 195:104-116. [PMID: 39870148 DOI: 10.1016/j.actbio.2025.01.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
The biomechanical properties of articular cartilage arise from a complex bioenvironment comprising hierarchically organised collagen networks within the extracellular matrix (ECM) that interact with the proteoglycan-rich interstitial fluid. This network features a depth-dependent fibril organisation across different zones. Understanding how collagen fibrils respond to external loading is key to elucidating the mechanisms behind lesion formation and managing degenerative conditions like osteoarthritis. This study employs polarisation-resolved second harmonic generation (pSHG) microscopy to quantify the ultrastructural organisation of collagen fibrils and their spatial gradient along the depth of bone-cartilage explants under a close-to-in vivo condition. By combining with in-situ loading, we examined the responses of collagen fibrils by quantifying changes in their principal orientation and degree of alignment. The spatial gradient and heterogeneity of collagen organisation were captured at high resolution (1 μm) along the longitudinal plane of explants (0.5 mm by 2 mm). Zone-specific ultrastructural characteristics were quantified to aid in defining zonal borders, revealing consistent zonal proportions with varying overall thicknesses. Under compression, the transitional zone exhibited the most significant re-organisation of collagen fibrils. It initially allowed large deformation through the re-orientation of fibrils, which then tightened fibril alignment to prevent excessive deformation, indicating a dynamic adaptation mechanism in response to increasing strain levels. Our results provide comprehensive, zone-specific baselines of cartilage ultrastructure and micromechanics, crucial for investigating the onset and progression of degenerative conditions, setting therapeutic intervention targets, and guiding cartilage repair and regeneration efforts. STATEMENT OF SIGNIFICANCE: Achieved unprecedented quantification of the spatial gradient and heterogeneity of collagen ultrastructural organisation at a high resolution (1 μm) along the full depth of the longitudinal plane of osteochondral explants (0.5 mm by 2 mm) under close-to-in vivo condition. Suggested new anatomical landmarks based on ultrastructural features for determining zonal borders and found consistent zonal proportions in explants with different overall thicknesses. Demonstrated that collagen fibrils initially respond by reorienting themselves at low strain levels, playing a significant role in cartilage deformation, particularly within the transitional zone. At higher strain levels, more collagen fibrils re-aligned, indicating a dynamic shift in the response mechanism at varying strain levels.
Collapse
Affiliation(s)
- Jingrui Hu
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, UK
| | - Keke Zheng
- Institute for Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, UK
| | - Benjamin E Sherlock
- Biomedical Physics, Faculty of Environment, Science and Economy, University of Exeter, UK
| | - Jingxiao Zhong
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, UK; School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, Australia
| | - Jessica Mansfield
- Biomedical Physics, Faculty of Environment, Science and Economy, University of Exeter, UK
| | - Ellen Green
- Biomedical Physics, Faculty of Environment, Science and Economy, University of Exeter, UK
| | - Andrew D Toms
- Exeter Knee Reconstruction Unit, Princess Elizabeth Orthopaedic Centre, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - C Peter Winlove
- Biomedical Physics, Faculty of Environment, Science and Economy, University of Exeter, UK
| | - Junning Chen
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, UK.
| |
Collapse
|
9
|
Xu S, Zhang J, Che J, Sha Z, Zhang M, Zhang Z, Xiang T, Zhong W, Wu L, Lv G, Cai B, Xu L, Xu C, Chen Z. Lysyl oxidase nanozyme-loaded hydrogel for sustained release and promotion of diabetic bone defect regeneration. J Control Release 2025; 379:927-943. [PMID: 39864630 DOI: 10.1016/j.jconrel.2025.01.070] [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: 08/07/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
The process of regenerating bone injuries in diabetic presents significant challenges because lysine oxidase (LOX), a key catalytic enzyme for collagen cross-linking, is inhibited in hyperglycemia. The supplementation of LOX is constrained by inadequate sources and diminished enzymatic activity, necessitating the development of effective alternatives for enhancing bone regeneration in diabetes. Herein, we reported a lysyl oxidase nanozyme (LON), derived from the catalytic domain of LOX. LON formed a stable coordination structure with the active center Cu2+ through histidine imidazolyl nitrogen and quinone oxygen, which is consistent with the conformation of the LOX. Our findings suggested that LON demonstrated the capacity to substitute LOX in promoting collagen synthesis and biomineralization. To enable sustained LON delivery, it was incorporated into a GelMA hydrogel (GH), forming a sustained-release reservoir known as LON-GelMA hydrogel (LONGH). Mechanism of LONGH promoting bone healing to accelerate the crosslinking and maturation stage of collagen were also explored, and the 23 genes closely associated with collagen regeneration and osteogenesis were found to be upregulated. The present investigation outcomes reveal that the engineered LONGH hydrogel presents a novel, simple, and commercially viable approach for bone regeneration, offering significant potential for clinical applications.
Collapse
Affiliation(s)
- Shan Xu
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China; Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214125, China
| | - Jian Zhang
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Jun Che
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Zhengzhou Sha
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Mingtao Zhang
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Zhiyan Zhang
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Tangyong Xiang
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Weixi Zhong
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Li Wu
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Gaohong Lv
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Baochang Cai
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Liu Xu
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| | - Cheng Xu
- Pharmacy department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215300, China.
| | - Zhipeng Chen
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| |
Collapse
|
10
|
Chen W, Meng J, Wang S. Bioinspired Materials for Controlling Mineral Adhesion: From Innovation Design to Diverse Applications. ACS NANO 2025; 19:7546-7582. [PMID: 39979232 DOI: 10.1021/acsnano.4c16946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The advancement of controllable mineral adhesion materials has significantly impacted various sectors, including industrial production, energy utilization, biomedicine, construction engineering, food safety, and environmental management. Natural biological materials exhibit distinctive and controllable adhesion properties that inspire the design of artificial systems for controlling mineral adhesion. In recent decades, researchers have sought to create bioinspired materials that effectively regulate mineral adhesion, significantly accelerating the development of functional materials across various emerging fields. Herein, we review recent advances in bioinspired materials for controlling mineral adhesion, including bioinspired mineralized materials and bioinspired antiscaling materials. First, a systematic overview of biological materials that exhibit controllable mineral adhesion in nature is provided. Then, the mechanism of mineral adhesion and the latest adhesion characterization between minerals and material surfaces are introduced. Later, the latest advances in bioinspired materials designed for controlling mineral adhesion are presented, ranging from the molecular level to micro/nanostructures, including bioinspired mineralized materials and bioinspired antiscaling materials. Additionally, recent applications of these bioinspired materials in emerging fields are discussed, such as industrial production, energy utilization, biomedicine, construction engineering, and environmental management, highlighting their roles in promoting or inhibiting aspects. Finally, we summarize the ongoing challenges and offer a perspective on the future of this charming field.
Collapse
Affiliation(s)
- Wei Chen
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jingxin Meng
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
11
|
Wu X, Yang H, Liu G, Sun W, Li J, Zhao Y, Gao X, Liu X, Song F, Wang S, Lu Z, Chen C, Huang C. Osteomimix: A Multidimensional Biomimetic Cascade Strategy for Bone Defect Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416715. [PMID: 39924776 DOI: 10.1002/adma.202416715] [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: 10/31/2024] [Revised: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Despite advancements in biomimetic mineralization techniques, the repair of large-scale bone defects remains a significant challenge. Inspired by the bone formation process, a multidimensional biomimetic cascade strategy is developed by replicating the biomineralization cascade, emulating the hierarchical structure of bone, and biomimicking its biological functions for efficient bone regeneration. This strategy involves the photocrosslinking of sodium methacrylate carboxymethyl cellulose-stabilized amorphous magnesium-calcium phosphate with methacrylate-modified type I collagen to create a self-mineralizing hydrogel. The hydrogel is then integrated with either naturally derived or synthetic oriented bulk scaffolds. The resulting composite, named Osteomimix, provides excellent mechanical support and can be customized for irregular bone defects using CAD/CAM technology. Through in vitro and in vivo studies, this work finds that Osteomimix exhibits spontaneous in situ biomimetic mineralization in a cell-free environment, while modulating immune responses and promoting vascularized bone formation in a cell-dependent manner. Built on bone-specific insights, this strategy achieves biomimicry across temporal, spatial, and functional dimensions, facilitating the seamless integration of artificial constructs with the natural tissue repair dynamics.
Collapse
Affiliation(s)
- Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Gufeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jiyun Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xin Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Shilei Wang
- Hubei Engineering Center of Natural Polymer-Based Medical Materials and Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ziyang Lu
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Chaoji Chen
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, School of Resource and Environmental Sciences, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
12
|
Zheng Z, Jiang M, Lv Z, Liu Y, Zhang H, Fang W, Xie H, Wang H, Wang W, Ping H, Li B, Fu Z. Enhanced Ion Transport Through Organized Cadmium Carbonate Nanocrystals in Collagen Fibrils for Efficient Biological Memristors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410205. [PMID: 39930705 DOI: 10.1002/smll.202410205] [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: 10/30/2024] [Revised: 01/24/2025] [Indexed: 03/20/2025]
Abstract
Owing to the unique assembly of collagen molecules, collagen fibrils have a confined structure that can effectively guide the intrafibrillar-oriented growth of inorganic crystals, such as hydroxyapatite and calcium carbonate. However, utilizing this organized structure of mineralized collagen fibrils for rapid ion transport remains challenging. Herein, the oriented growth of functional cadmium carbonate (CdCO3) nanocrystals is reported within collagen fibrils and demonstrates that different areas within a single mineralized collagen fibril exhibit a uniform orientation. The results show that the precursor phase infiltrates the collagen through the gap zones owing to collagen confinement, gradually transforming into well-oriented crystalline nanocrystals within the collagen. Adopting the principles of intrafibrillar mineralization with CdCO3, the mineralization process of collagen matrices can be regulated, such as collagen films and tendon slices, by adjusting the mineralization temperature, thereby modulating the stress generated in the collagen matrices, thus highlighting new possibilities for using organized biominerals in rapid ion transport. Additionally, the use of mineralized collagen fibrils are demonstrated in biological memristors. The fabricated memristor device exhibits a low set voltage (0.65 V) and high on/off ratio (≈106), highlighting the potential of mineralized collagen in advanced electronic applications.
Collapse
Affiliation(s)
- Zhuozhi Zheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China
| | - Minghao Jiang
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Ziyu Lv
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Yin Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Huanhuan Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Weijian Fang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Hao Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Weimin Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| | - Hang Ping
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China
| | - Bin Li
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China
| | - Zhengyi Fu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Luoshi Road No. 122, Wuhan, 430070, China
| |
Collapse
|
13
|
Liu F, Hu Y, Zhang Y, Ren C, Qiao F, Yang H, Xu H, Yang P. Aging-Induced Discrepant Response of Fracture Healing is Initiated from the Organization and Mineralization of Collagen Fibrils in Callus. ACS Biomater Sci Eng 2025; 11:1038-1050. [PMID: 39831893 DOI: 10.1021/acsbiomaterials.4c01490] [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: 01/22/2025]
Abstract
Fracture healing is a complex process during which the bone restores its structural and mechanical integrity. Collagen networks and minerals are the fundamental components to rebuild the bone matrix in callus. It has been recognized that bone quality could be impaired during aging. However, how the structural and mechanical recovery of fracture healing is influenced by aging, particularly from the perspective of organization and mineralization of the collagen network in callus, remains unclear. A tibial fracture model was established for both the young (5 weeks) and aged mice (68 weeks). On the 21st day postfracture, the characteristics of the collagen network, mineralization, and the nanoscale mechanical properties of the callus were assessed. The results indicated that aging postpones the fracture healing process, leading to incomplete microstructure, less mineral content and mineralization, and weaker mechanical properties of callus. In the aged mice, the internal fixation and mechanical immobilization promoted the mineralization of callus by increasing mineral crystal length and mineral-to-matrix ratio by 48 and 42% compared to the internal fixation and free movement control group, respectively. By contrast, in the young mice, the internal fixation and mechanical immobilization induced disordered collagen fibrils and decreased the crystal length and mineral-to-matrix ratio by 32 and 36%, compared to the internal fixation and free movement control group, respectively. The present findings suggested that the aging-induced structure and mechanical differences of callus during fracture healing initiate from the organization and mineralization of collagen fibrils. Multiscale structural and mechanical analysis suggested mechanical immobilization is beneficial to the structure, composition, and mechanics of callus in the aged mice while impairing the organization and mineralization of collagen fibril in the callus of the young mice. These findings suggested that different mechanical intervention strategies should be adopted for fracture healing at different ages, which provides valuable insights for the clinical treatment of bone fracture.
Collapse
Affiliation(s)
- Fa Liu
- Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Ningbo 315103, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yiwei Hu
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yuzhi Zhang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chenxi Ren
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Feng Qiao
- Department of Orthopedics Combined TCM with Western Medicine, Honghui Hospital, Xi'an Jiaotong University, No. 555 Youyi East Road, Xi'an, Shaanxi 710054, China
| | - Hui Yang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Huiyun Xu
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Pengfei Yang
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
14
|
Han B, Song Y, Wang S, Yang T, Sun Z, Wang A, Jin M, Yang Z, Wang X, Liang F. Biomimetic Janus Particles Induced In Situ Interfacial Remineralization for Dentin Hypersensitivity. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202412954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Indexed: 02/03/2025]
Abstract
AbstractDentin hypersensitivity (DH), caused by the exposure of dentin tubules, is a common complaint of dental patients. Although occlusion of the exposed tubules is the primary treatment approach, the complex oral environment, and multiple simultaneous requirements often hinder its implementation. In this study, strawberry‐shaped hemispheric Janus particles (JPs) are synthesized, and their use in the treatment of DH is evaluated in vitro and in an animal model. The hemispheric side of the JPs is modified with polymers of quaternary ammonium salts (QASs) to form a superhydrophobic coating with antibiofilm properties, while the flat side is modified with catechol groups able to form strong bonds with dentin. Even after 1 h of ultrasonication or 1000 rounds of thermal cycling, the dentin tubules are completely occluded by the JPs. Moreover, biofilm formation is not observed, and the area of living bacteria is less than 1% compared to the blank control and sodium fluoride (NaF)‐treated groups. In a rat model, the dentin tubules in the fixed specimens are completely occluded at day 3, much earlier than the occlusion obtained with commonly used NaF. These results demonstrate that JPs can provide a novel approach to the treatment of DH.
Collapse
Affiliation(s)
- Bing Han
- Department of Cariology and Endodontology Peking University School and Hospital of Stomatology National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology Beijing 100081 P. R. China
| | - Yilin Song
- Department of Cariology and Endodontology Peking University School and Hospital of Stomatology National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology Beijing 100081 P. R. China
| | - Shi Wang
- Institute of Polymer Science and Engineering Department of Chemical Engineering Tsinghua University Beijing 100084 P. R. China
| | - Tiantian Yang
- School of Environmental and Chemical Engineering Shenyang University of Technology Shenyang 110870 P. R. China
| | - Zetao Sun
- Institute of Polymer Science and Engineering Department of Chemical Engineering Tsinghua University Beijing 100084 P. R. China
| | - Aijing Wang
- Department of Cariology and Endodontology Peking University School and Hospital of Stomatology National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology Beijing 100081 P. R. China
| | - Moran Jin
- Department of Cariology and Endodontology Peking University School and Hospital of Stomatology National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology Beijing 100081 P. R. China
| | - Zhenzhong Yang
- Institute of Polymer Science and Engineering Department of Chemical Engineering Tsinghua University Beijing 100084 P. R. China
| | - Xiaoyan Wang
- Department of Cariology and Endodontology Peking University School and Hospital of Stomatology National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology Beijing 100081 P. R. China
| | - Fuxin Liang
- Institute of Polymer Science and Engineering Department of Chemical Engineering Tsinghua University Beijing 100084 P. R. China
| |
Collapse
|
15
|
Wang M, Jiang M, Wang Q, Sun Y, Nie Z, Palin WM, Zhang Z. An in vitro bioinspired approach to enhance the bioactivity of titanium implants via electrophoretic deposition and biomimetic mineralization of type i collagen. BIOMATERIALS ADVANCES 2025; 167:214110. [PMID: 39541891 DOI: 10.1016/j.bioadv.2024.214110] [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: 07/10/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study aims to explore the efficacy of Electrophoretic Deposition (EPD) for collagen type I coating on titanium implants and its subsequent mineralization to improve osseointegration and bone regeneration. METHODS Titanium disks were prepared with a sandblasted, large grit and acid-etched (SLA) surface. EPD was employed to deposit collagen type I onto the titanium surfaces, followed by two modes of mineralization: extra-fibril mineralization (EFM) and inter-fibril mineralization (IFM). Then comprehensive in vitro studies were conducted including surface properties, cell proliferation, osteogenic differentiation, and inflammatory responses. RESULTS EPD successfully deposited a uniform collagen layer on titanium surfaces. EFM resulted in deposition of larger, irregularly shaped crystals, while IFM produced controlled, helical fibril mineralization. IFM-treated surfaces exhibited enhanced cell viability, proliferation, and osteogenic differentiation. Both EFM and IFM surfaces triggered higher macrophage activation than SLA surfaces. While EFM primarily induced a stronger M1 pro-inflammatory response, IFM exhibited a more balanced macrophage polarization with upregulated M2 markers at later stages. CONCLUSION EPD, particularly when integrated with IFM, significantly enhances the bioactivity and osteogenic potential of collagen-coated titanium implants. This method surpasses traditional SLA surfaces by stabilizing the collagen layer and creating a biomimetic environment conducive to bone regeneration and healing through a balanced inflammatory response, offering a promising strategy to improve titanium implant performance.
Collapse
Affiliation(s)
- Man Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Muqi Jiang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qi Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yasheng Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Zhixiang Nie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - William M Palin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Dental and Biomaterials Sciences, School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, UK.
| | - Zhen Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Dental and Biomaterials Sciences, School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, UK; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
16
|
Liu Y, Gao X, Li Y, Gao A, Zheng Z, Wei J, Yang H, Ping H, Xie H, Wang H, Wang W, Fu Z. Intrafibrillar calcium carbonate mineralization of electrospinning polyvinyl alcohol/collagen films with improved mechanical and bioactive properties. J Mater Chem B 2024; 13:312-325. [PMID: 39540843 DOI: 10.1039/d4tb01472c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Collagen films play an essential role in guided bone-regeneration (GBR) techniques, which create space, promote cell adhesion, and induce osteogenic differentiation. It is therefore crucial to design appropriate GBR films to facilitate bone regeneration. However, current electrospun collagen scaffolds used as bioactive materials have limited clinical applications due to their poor mechanical properties. In this study, polyvinyl alcohol (PVA)/collagen (Col) films were electrospun by mixing PVA and type I collagen solution. For the first time, the intrafibrillar mineralization of aragonite nanocrystals within the PVA/Col fibrils was achieved, resulting in the formation of a hierarchical, bioactive film. The PVA/Col-CaCO3 film exhibited good mechanical properties, with hardness and Young's modulus values of 211.6 ± 0.1 MPa and 5.6 ± 1.7 GPa, respectively. Furthermore, bone marrow mesenchymal stem cells (BMSCs) inoculated onto the PVA/Col-CaCO3 film demonstrated robust adhesion and proliferation. The mineralized fibrils effectively stimulated the growth of BMSCs while suppressing cell apoptosis. Besides, the PVA/Col-CaCO3 film significantly induced the osteogenic differentiation of BMSCs, revealing its potential biomedical applications in hard tissue engineering.
Collapse
Affiliation(s)
- Yin Liu
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China.
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Xin Gao
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430070, China
| | - Yuqi Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, P. R. China
| | - Anqi Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhuozhi Zheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Jingjiang Wei
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China.
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430070, China
| | - Hang Ping
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China.
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Hao Xie
- School of Chemistry, Chemical Engineering, and Life Science Wuhan University of Technology, Wuhan, 430070, China
| | - Hao Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Weimin Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhengyi Fu
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang, 441000, China.
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| |
Collapse
|
17
|
Liu C, Xie Y, Zhang Y, Sun Y, Wang Y, Han B, Jiang X, Wang B. MOF-Based Guided Bone Regeneration Membrane for Promoting Osteogenesis by Regulating Bone Microenvironment through Cascade Effects. Adv Healthc Mater 2024:e2403187. [PMID: 39632369 DOI: 10.1002/adhm.202403187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Regulation of bone microenvironment (BME) including innate pH values and metal ions affects cellular functions and activities of osteoblasts and osteoclasts, thereby significantly influencing the process of bone regeneration. How to achieve multiple effective regulations of the BME through cascade effects via facile material design and fabrication to significantly facilitate osteogenesis remains a challenge. Herein, a facilely-designed resorbable guided bone regeneration membrane (PCL/DEX@Ca-Zol) based on a drug-loaded metal-organic framework is reported. Thereinto, calcium ions, zoledronic acid, and dexamethasone embedded in the membrane can be responsively released specifically inside bone defect in an acid-triggered manner to synergistically regulate BME by neutralization of pH value, enhancement of osteogenic differentiation and mineralization, and inhibition of osteoclasts in one-go. Along with polycaprolactone as a structural support in the membrane for bone regeneration with fully utilized components of the composite membrane material, enhances bone regeneration with minimized side effects is accordingly achieved with the assistance of effective modulation of BME through multiple cascade effects.
Collapse
Affiliation(s)
- Chunyu Liu
- Frontiers Science Centre for High Energy Material, Advanced Technology Research Institute (Jinan), Key Laboratory of Cluster Science (Ministry of Education), Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Yajuan Xie
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yunfan Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Yuqi Sun
- Frontiers Science Centre for High Energy Material, Advanced Technology Research Institute (Jinan), Key Laboratory of Cluster Science (Ministry of Education), Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Yuanbo Wang
- Frontiers Science Centre for High Energy Material, Advanced Technology Research Institute (Jinan), Key Laboratory of Cluster Science (Ministry of Education), Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Bing Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, P. R. China
| | - Xin Jiang
- Department of Orthopedics and Spine Surgery, China-Japan Friendship Hospital, Beijing, 100029, P. R. China
| | - Bo Wang
- Frontiers Science Centre for High Energy Material, Advanced Technology Research Institute (Jinan), Key Laboratory of Cluster Science (Ministry of Education), Beijing Key Laboratory of Photoelectronic/Electrophotonic Conversion Materials, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
18
|
Misof BM, Fratzl-Zelman N. Bone Quality and Mineralization and Effects of Treatment in Osteogenesis Imperfecta. Calcif Tissue Int 2024; 115:777-804. [PMID: 39231826 DOI: 10.1007/s00223-024-01263-8] [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: 03/21/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024]
Abstract
Osteogenesis imperfecta (OI) is a rare congenital bone dysplasia characterized by high fracture rates and broad variations in clinical manifestations ranging from mild to increasingly severe and perinatal lethal forms. The underlying mutations affect either the synthesis or processing of the type I procollagen molecule itself or proteins that are involved in the formation and mineralization of the collagen matrix. Consequently, the collagen forming cells, the osteoblasts, become broadly dysfunctional in OI. Strikingly, hypermineralized bone matrix seems to be a frequent feature in OI, despite the variability in clinical severity and mutations in the so far studied different forms of human OI. While the causes of the increased mineral content of the bone matrix are not fully understood yet, there is evidence that the descendants of the osteoblasts, the osteocytes, which play a critical role not only in bone remodeling, but also in mineralization and sensing of mechanical loads, are also highly dysregulated and might be of major importance in the pathogenesis of OI. In this review article, we firstly summarize findings of cellular abnormalities in osteoblasts and osteocytes, alterations of the organic matrix, as well as of the microstructural organization of bone. Secondly, we focus on the hypermineralization of the bone matrix in OI as observed in several different forms of human OI as well as in animal models, its measurement and potential mechanical implications and its effect on the bone mineral density measured by dual X-ray absorptiometry. Thirdly, we give an overview of established medication treatments of OI and new approaches with a focus of their known or possible effects on the bone material, particularly on bone matrix mineralization.
Collapse
Affiliation(s)
- Barbara M Misof
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Med. Dept. Hanusch Hospital, Vienna, Austria
- Vienna Bone and Growth Center, Vienna, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Med. Dept. Hanusch Hospital, Vienna, Austria.
- Vienna Bone and Growth Center, Vienna, Austria.
| |
Collapse
|
19
|
Schemenz V, Scoppola E, Zaslansky P, Fratzl P. Bone strength and residual compressive stress in apatite crystals. J Struct Biol 2024; 216:108141. [PMID: 39442775 DOI: 10.1016/j.jsb.2024.108141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Residual stresses are omnipresent in composite materials, often arising during the fabrication process. Residual compressive stresses were recently observed to develop in collagen fibrils during the process of mineralization. They have in fact been reported in a range of bony materials spanning tooth dentin to mammalian and fish bones. Treatment by heat or by irradiation have shown that compressive residual stresses up to 100 MPa can be released in the mineral by inducing damage to the protein fibers. This mini-review assembles some of the knowledge about residual stresses in bony nanocomposites and uses a composite model to argue that such stresses play a major role in enhancing the strength of bone.
Collapse
Affiliation(s)
- Victoria Schemenz
- Department for Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Germany
| | - Ernesto Scoppola
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Paul Zaslansky
- Department for Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Germany
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany.
| |
Collapse
|
20
|
Nutan B, Okada M, Matsumoto T. Lipids and Minerals, Interplay in Biomineralization: Nature's Alchemy. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:571-580. [PMID: 38386501 DOI: 10.1089/ten.teb.2023.0249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The main focus of this article is the role of lipids in biomineralization. Much of the discussion on biomineralization focuses on proteins in these decades. Indeed, collagen and acidic noncollagenous proteins effectively serve as templates for mineralization. However, other macromolecules such as lipids and polysaccharides have received less attention despite their abundance at mineralization sites. The matrix vesicle (MV) theory is widely accepted as the induction of early mineralization. Although ion concentration within the vesicles has been discussed in the initial mineralization in this theory, the role of phospholipids that constitute the vesicle membrane has not been discussed much. Comprehensive considerations, including pathological mineralization, exist regardless of the localization of MVs, the involvement of bacteria in dental calculus formation, and biomineralization caused by marine organisms such as corals, suggesting that initial mineralization found in these biological conditions might be a common reaction relating to lipids. In contrast, despite the abundance of lipids, mineralization occurs only in the limited tissue within our body. In other words, gathering knowledge and creating a path to understanding about lipid-based mineralization is extremely important in proposing new bone disease treatment methods. This article describes how lipids influence nucleation, mineralization, and expansion during hard tissue formation. Impact statement Recent studies have accumulated evidence of mineralization involving phospholipids and the matrix vesicle (MV) theory. Mineralization occurs not only in the conventional vesicle form but also in flat membranes arrested by the matrix. The flat membrane is derived not only from MVs but also from various causes, such as cell rupture and cell apoptosis. Mineralization is greatly affected by alkaline phosphatases derived from cell membranes. By understanding phospholipid-based mineralization, it will be possible to design new mineralization-inducing materials centered on cellular components for early bone formation. This article is important for developing new strategies to induce bone mineralization.
Collapse
Affiliation(s)
- Bhingaradiya Nutan
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Okada
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
21
|
Bai X, Peng W, Tang Y, Wang Z, Guo J, Song F, Yang H, Huang C. An NIR-propelled janus nanomotor with enhanced ROS-scavenging, immunomodulating and biofilm-eradicating capacity for periodontitis treatment. Bioact Mater 2024; 41:271-292. [PMID: 39149593 PMCID: PMC11324457 DOI: 10.1016/j.bioactmat.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 08/17/2024] Open
Abstract
Periodontitis is an inflammatory disease caused by bacterial biofilms, which leads to the destruction of periodontal tissue. Current treatments, such as mechanical cleaning and antibiotics, struggle to effectively address the persistent biofilms, inflammation, and tissue damage. A new approach involves developing a Janus nanomotor (J-CeM@Au) by coating cerium dioxide-doped mesoporous silica (CeM) with gold nanoparticles (AuNPs). This nanomotor exhibits thermophoretic motion when exposed to near-infrared (NIR) laser light due to the temperature gradient produced by the photothermal effects of asymmetrically distributed AuNPs. The NIR laser provides the energy for propulsion and activates the nanomotor's antibacterial properties, allowing it to penetrate biofilms and kill bacteria. Additionally, the nanomotor's ability to scavenge reactive oxygen species (ROS) can modulate the immune response and create a regenerative environment, promoting the healing of periodontal tissue. Overall, this multifunctional nanomotor offers a promising new approach for treating periodontitis by simultaneously addressing biofilm management and immune modulation with autonomous movement.
Collapse
Affiliation(s)
- Xuan Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wenan Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Ying Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Ziming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jingmei Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
22
|
Yi J, Li M, Zhu J, Wang Z, Li X. Recent development and applications of electrodeposition biocoatings on medical titanium for bone repair. J Mater Chem B 2024; 12:9863-9893. [PMID: 39268681 DOI: 10.1039/d4tb01081g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Bioactive coatings play a crucial role in enhancing the osseointegration of titanium implants for bone repair. Electrodeposition offers a versatile and efficient technique to deposit uniform coatings onto titanium surfaces, endowing implants with antibacterial properties, controlled drug release, enhanced osteoblast adhesion, and even smart responsiveness. This review summarizes the recent advancements in bioactive coatings for titanium implants used in bone repair, focusing on various electrodeposition strategies based on material-structure synergy. Firstly, it outlines different titanium implant materials and bioactive coating materials suitable for bone repair. Then, it introduces various electrodeposition methods, including electrophoretic deposition, anodization, micro-arc oxidation, electrochemical etching, electrochemical polymerization, and electrochemical deposition, discussing their applications in antibacterial, osteogenic, drug delivery, and smart responsiveness. Finally, it discusses the challenges encountered in the electrodeposition of coatings for titanium implants in bone repair and potential solutions.
Collapse
Affiliation(s)
- Jialong Yi
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Ming Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Jixiang Zhu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - ZuHang Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Xiaoyan Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
23
|
Kuang Q, Feng S, Yang M. Biomimetic Aramid Nanofiber/β-FeOOH Composite Coating for Polypropylene Separators in Lithium-Sulfur Batteries. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39358833 DOI: 10.1021/acsami.4c10381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Aramid nanofibers (ANFs), with attractive mechanical and thermal properties, have attracted much attention as key building units for the design of high-performance composite materials. Although great progress has been made, the potential of ANFs as fibrous protein mimetics for controlling the growth of inorganic materials has not been fully revealed, which is critical for avoiding phase separation associated with typical solution blending. In this work, we show that ANFs could template the oriented growth of β-FeOOH nanowhiskers, which enables the synthesis of ANFs/β-FeOOH hybrids as composite coatings for polypropylene (PP) separators in Li-S batteries. The modified PP separator exhibits enhanced mechanical properties, heightened thermal performance, optimized electrolyte wettability, and improved ion conductivity, leading to superior electrochemical properties, including high initial specific capacity, better rate capability, and long cycling stability, which are superior to those of the commercial PP separators. Importantly, the addition of β-FeOOH to ANFs could further contribute to the suppression of lithium polysulfide shuttling by chemical immobilization, inhibition of the growth of lithium dendrites because of the intrinsic high modulus and hardness, and promotion of reaction dynamics due to the catalytic effect. We believe that our work may provide a potent biomimetic pathway for the development of advanced battery separators based on ANFs.
Collapse
Affiliation(s)
- Qingxia Kuang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Ming Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
24
|
Gao Q, Jiang Y, Zhou D, Li G, Han Y, Yang J, Xu K, Jing Y, Bai L, Geng Z, Zhang H, Zhou G, Zhu M, Ji N, Han R, Zhang Y, Li Z, Wang C, Hu Y, Shen H, Wang G, Shi Z, Han Q, Chen X, Su J. Advanced glycation end products mediate biomineralization disorder in diabetic bone disease. Cell Rep Med 2024; 5:101694. [PMID: 39173634 PMCID: PMC11524989 DOI: 10.1016/j.xcrm.2024.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 06/04/2024] [Accepted: 07/26/2024] [Indexed: 08/24/2024]
Abstract
Patients with diabetes often experience fragile fractures despite normal or higher bone mineral density (BMD), a phenomenon termed the diabetic bone paradox (DBP). The pathogenesis and therapeutics opinions for diabetic bone disease (DBD) are not fully explored. In this study, we utilize two preclinical diabetic models, the leptin receptor-deficient db/db mice (DB) mouse model and the streptozotocin-induced diabetes (STZ) mouse model. These models demonstrate higher BMD and lower mechanical strength, mirroring clinical observations in diabetic patients. Advanced glycation end products (AGEs) accumulate in diabetic bones, causing higher non-enzymatic crosslinking within collagen fibrils. This inhibits intrafibrillar mineralization and leads to disordered mineral deposition on collagen fibrils, ultimately reducing bone strength. Guanidines, inhibiting AGE formation, significantly improve the microstructure and biomechanical strength of diabetic bone and enhance bone fracture healing. Therefore, targeting AGEs may offer a strategy to regulate bone mineralization and microstructure, potentially preventing the onset of DBD.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China.
| | - Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yafei Han
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Jingzhi Yang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, 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
| | - Guangyin Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Mengru Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Ruina Han
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, Shanghai University, Shanghai 200444, P.R. China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, P.R. China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Chuandong Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Hao Shen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Zhongmin Shi
- Department of Orthopedics, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Qinglin Han
- Orthopaedic Department, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China.
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China; Organoid Research Center, 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
|
25
|
Wang J, Cui W. Decoding the diabetic bone paradox: How AGEs sabotage skeletal integrity. Cell Rep Med 2024; 5:101693. [PMID: 39293394 PMCID: PMC11524976 DOI: 10.1016/j.xcrm.2024.101693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/20/2024]
Abstract
Diabetes patients often suffer from fractures despite normal or high bone mineral density, a phenomenon known as the diabetic bone paradox. Gao et al.1 identify AGEs as disrupting bone quality and compromising skeletal integrity in diabetic bone disease.
Collapse
Affiliation(s)
- Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P.R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P.R. China.
| |
Collapse
|
26
|
Cheng Y, Yang Y, Wang S, Zhou Z, Li J, Zhang Y, Chen S, Zeng Z, Xie S, Tang BZ. Fluorogenic in-situ Labelling of Gelatin Polymer in Aqueous Solution and Hydrogel. Chemistry 2024; 30:e202401561. [PMID: 38847762 DOI: 10.1002/chem.202401561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Indexed: 08/31/2024]
Abstract
Gelatin polymers made from partially degraded collagen are important biomaterials, but their in-situ analysis suffers from uncontrollable covalent labelling and poor spatial-temporal imaging resolution. Herein, three tetrazolate-tagged tetraphenylethylene fluorophores (TPE-TAs) are introduced for practical fluorogenic labelling of gelatin in aqueous phase and hydrogels. These probes with aggregation-induced emission characteristics offer negligible background and elicit turn-on fluorescence by simply mixing with the gelatin in aqueous phase, giving a detection limit of 0.15 mg/L over a linear dynamic range up to 100 mg/L. This method does not work for collagens and causes minimal interference with gelatin properties. Mechanistic studies reveal a key role for multivalent electrostatic interactions between the abundant basic residues in gelatin (e. g., lysine, hydroxylysine, arginine) and anionic tetrazolate moieties of the lipophilic fluorophore synergistically in spatially rigid macromolecular encapsulation to achieve fluorogenic labelling. The AIE strategy by forming non-covalent fluorophore-gelatin complexes was developed for novel hydrogels that exhibited reversible fluorescence in response to dynamic microstructural changes in the hydrogel scaffold upon salting-in/out treatments, and enabled high spatial-temporal imaging of the fiber network in lyophilized samples. This work may open up avenues for in-situ imaging analysis and evaluation of gelatin-based biomaterials during processes such as in vivo degradation and mineralization.
Collapse
Affiliation(s)
- Yao Cheng
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yujiao Yang
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shuodong Wang
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhibiao Zhou
- School of Life Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiangcan Li
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yang Zhang
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Sijie Chen
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, China
| | - Zebing Zeng
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Sheng Xie
- Shenzhen Research Institute of Hunan University, Nanshan District, Shenzhen, 518000, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ben Zhong Tang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, 518172, China
| |
Collapse
|
27
|
Micheletti C, Shah FA. Bone hierarchical organization through the lens of materials science: Present opportunities and future challenges. Bone Rep 2024; 22:101783. [PMID: 39100913 PMCID: PMC11295937 DOI: 10.1016/j.bonr.2024.101783] [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: 12/31/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Multiscale characterization is essential to better understand the hierarchical architecture of bone and an array of analytical methods contributes to exploring the various structural and compositional aspects. Incorporating X-ray tomography, X-ray scattering, vibrational spectroscopy, and atom probe tomography alongside electron microscopy provides a comprehensive approach, offering insights into the diverse levels of organization within bone. X-ray scattering techniques reveal information about collagen-mineral spatial relationships, while X-ray tomography captures 3D structural details, especially at the microscale. Electron microscopy, such as scanning and transmission electron microscopy, extends resolution to the nanoscale, showcasing intricate features such as collagen fibril organization. Additionally, atom probe tomography achieves sub-nanoscale resolution and high chemical sensitivity, enabling detailed examination of bone composition. Despite various technical challenges, a correlative approach allows for a comprehensive understanding of bone material properties. Real-time investigations through in situ and in operando approaches shed light on the dynamic processes in bone. Recently developed techniques such as liquid, in situ transmission electron microscopy provide insights into calcium phosphate formation and collagen mineralization. Mechanical models developed in the effort to link structure, composition, and function currently remain oversimplified but can be improved. In conclusion, correlative analytical platforms provide a holistic perspective of bone extracellular matrix and are essential for unraveling the intricate interplay between structure and composition within bone.
Collapse
Affiliation(s)
- Chiara Micheletti
- Department of Biomaterials, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Furqan A. Shah
- Department of Biomaterials, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
Grünewald TA, Liebi M, Birkedal H. Crossing length scales: X-ray approaches to studying the structure of biological materials. IUCRJ 2024; 11:708-722. [PMID: 39194257 PMCID: PMC11364038 DOI: 10.1107/s2052252524007838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Biological materials have outstanding properties. With ease, challenging mechanical, optical or electrical properties are realised from comparatively `humble' building blocks. The key strategy to realise these properties is through extensive hierarchical structuring of the material from the millimetre to the nanometre scale in 3D. Though hierarchical structuring in biological materials has long been recognized, the 3D characterization of such structures remains a challenge. To understand the behaviour of materials, multimodal and multi-scale characterization approaches are needed. In this review, we outline current X-ray analysis approaches using the structures of bone and shells as examples. We show how recent advances have aided our understanding of hierarchical structures and their functions, and how these could be exploited for future research directions. We also discuss current roadblocks including radiation damage, data quantity and sample preparation, as well as strategies to address them.
Collapse
Affiliation(s)
| | - Marianne Liebi
- Photon Science DivisionPaul Scherrer InstituteVilligenPSI5232Switzerland
- Institute of MaterialsÉcole Polytechnique Fédérale de Lausanne1015 LausanneSwitzerland
| | - Henrik Birkedal
- Department of Chemistry & iNANOAarhus UniversityGustav Wieds Vej 14Aarhus8000Denmark
| |
Collapse
|
29
|
Rong M, Liu D, Xu X, Li A, Bai Y, Yang G, Liu K, Zhang Z, Wang L, Wang K, Lu L, Jiang Y, Liu J, Zhang X. A Superparamagnetic Composite Hydrogel Scaffold as In Vivo Dynamic Monitorable Theranostic Platform for Osteoarthritis Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405641. [PMID: 38877353 DOI: 10.1002/adma.202405641] [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: 04/20/2024] [Revised: 05/27/2024] [Indexed: 06/16/2024]
Abstract
Osteoarthritis (OA) is a prevalent disease, characterized by subchondral fractures in its initial stages, which has no precise and specific treatment now. Here, a novel multifunctional scaffold is synthesized by photopolymerizing glycidyl methacrylate-modified hyaluronic acid (GMHA) as the matrix in the presence of hollow porous magnetic microspheres based on hydroxyapatite. In vivo subchondral bone repairing results demonstrate that the scaffold's meticulous design has most suitable properties for subchondral bone repair. The porous structure of inorganic particles within the scaffold facilitates efficient transport of loaded exogenous vascular endothelial growth factor (VEGF). The Fe3O4 nanoparticles assembled in microspheres promote the osteogenic differentiation of bone marrow mesenchymal stem cells and accelerate the new bone generation. These features enable the scaffold to exhibit favorable subchondral bone repair properties and attain high cartilage repair scores. The therapy results prove that the subchondral bone support considerably influences the upper cartilage repair process. Furthermore, magnetic resonance imaging monitoring demonstrates that Fe3O4 nanoparticles, which are gradually replaced by new bone during osteochondral defect repair, allow a noninvasive and radiation-free assessment to track the newborn bone during the OA repair process. The composite hydrogel scaffold (CHS) provides a versatile platform for biomedical applications in OA treatment.
Collapse
Affiliation(s)
- Mayifei Rong
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Dingge Liu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoguang Xu
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Ang Li
- Faculty of Materials and Manufacturing, Beijing Key Lab of Microstructure and Properties of Advanced Materials, Beijing University of Technology, Beijing, 100124, China
| | - Yihua Bai
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Gang Yang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Kaiping Liu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Zhihua Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Langran Wang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Kai Wang
- School of Mathematics and Physics, Handan University, Handan, 056005, China
| | - Liying Lu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yong Jiang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Ji Liu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xin Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
30
|
Yang Q, Zheng W, Zhao Y, Shi Y, Wang Y, Sun H, Xu X. Advancing dentin remineralization: Exploring amorphous calcium phosphate and its stabilizers in biomimetic approaches. Dent Mater 2024; 40:1282-1295. [PMID: 38871525 DOI: 10.1016/j.dental.2024.06.013] [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: 03/29/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE This review elucidates the mechanisms underpinning intrafibrillar mineralization, examines various amorphous calcium phosphate (ACP) stabilizers employed in dentin's intrafibrillar mineralization, and addresses the challenges encountered in clinical applications of ACP-based bioactive materials. METHODS The literature search for this review was conducted using three electronic databases: PubMed, Web of Science, and Google Scholar, with specific keywords. Articles were selected based on inclusion and exclusion criteria, allowing for a detailed examination and summary of current research on dentin remineralization facilitated by ACP under the influence of various types of stabilizers. RESULTS This review underscores the latest advancements in the role of ACP in promoting dentin remineralization, particularly intrafibrillar mineralization, under the regulation of various stabilizers. These stabilizers predominantly comprise non-collagenous proteins, their analogs, and polymers. Despite the diversity of stabilizers, the mechanisms they employ to enhance intrafibrillar remineralization are found to be interrelated, indicating multiple driving forces behind this process. However, challenges remain in effectively designing clinically viable products using stabilized ACP and maximizing intrafibrillar mineralization with limited materials in practical applications. SIGNIFICANCE The role of ACP in remineralization has gained significant attention in dental research, with substantial progress made in the study of dentin biomimetic mineralization. Given ACP's instability without additives, the presence of ACP stabilizers is crucial for achieving in vitro intrafibrillar mineralization. However, there is a lack of comprehensive and exhaustive reviews on ACP bioactive materials under the regulation of stabilizers. A detailed summary of these stabilizers is also instrumental in better understanding the complex process of intrafibrillar mineralization. Compared to traditional remineralization methods, bioactive materials capable of regulating ACP stability and controlling release demonstrate immense potential in enhancing clinical treatment standards.
Collapse
Affiliation(s)
- Qingyi Yang
- Department of Periodontology, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Wenqian Zheng
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yuping Zhao
- Department of Periodontology, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yaru Shi
- Department of Periodontology, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yi Wang
- Graduate Program in Applied Physics, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Xiaowei Xu
- Department of Periodontology, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
31
|
Liu Y, Song Y, Wu P. Self-Evolving Hierarchical Hydrogel Fibers with Circadian Rhythms and Memory Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404506. [PMID: 38837474 DOI: 10.1002/adma.202404506] [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: 03/28/2024] [Revised: 05/30/2024] [Indexed: 06/07/2024]
Abstract
The fusion of hierarchical tissues at interfaces, incorporating ultrafast selective transport channels, enables efficient matter exchange and energy transfer across multiscale structures in living organisms. However, achieving these characteristics simultaneously in an artificial multimaterial system is challenging. Here, this work presents a multimaterial hydrogel fiber with a hierarchical structure of interface fusion, which forms spontaneously through in situ hierarchy evolution induced by ionic cross-linking and molecular shear flow. Water transport occurs in the angstrom-scale confined slits created by aligned cellulose nanocrystals (CNCs) by direct Coulomb knock-on, resembling Newton's cradle motion. The fusion of interfaces enables high-efficiency water transport across multiscale layers, combined with Newton's cradle-like collective water motion, creating a highly sensitive negative feedback loop within the fiber. These fibers exhibit integrated behaviors of time-space perception, short-term memory and adaptive changes in shape. Additionally, they demonstrate rhythm characteristics, changing periodically in a 24-h day-night cycle. Composed of natural building blocks, these hierarchical hydrogel fibers exhibit a memristor effect similar to that of an elementary neuron, making them promising for applications in seamless on-skin and implantable bioelectronics.
Collapse
Affiliation(s)
- Yanjun Liu
- State Key Laboratory for Modification of Chemical Fibres and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Yuanzhu Song
- State Key Laboratory for Modification of Chemical Fibres and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Peiyi Wu
- State Key Laboratory for Modification of Chemical Fibres and Polymer Materials, College of Chemistry and Chemical Engineering, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| |
Collapse
|
32
|
Bai Y, Wu P, Zhang Q, Lin F, Hu L, Zhang Z, Huang W, Xiao Y, Zuo Q. Decorin in the spatial control of collagen mineralization. MATERIALS HORIZONS 2024; 11:3396-3407. [PMID: 38690683 DOI: 10.1039/d3mh02216a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Understanding the molecular mechanism by which the periodontal ligament (PDL) is maintained uncalcified between two mineralized tissues (cementum and bone) may facilitate the functional repair and regeneration of the periodontium complex, disrupted in the context of periodontal diseases. However, research that explores the control of type I collagen (COL I) mineralization fails to clarify the detailed mechanism of regulating spatial collagen mineralization, especially in the periodontium complex. In the present study, decorin (DCN), which is characterized as abundant in the PDL region and rare in mineralized tissues, was hypothesized to be a key regulator in the spatial control of collagen mineralization. The circular dichroism results confirmed that DCN regulated the secondary structure of COL I, and the surface plasmon resonance results indicated that COL I possessed a higher affinity for DCN than for other mineralization promoters, such as DMP-1, OPN, BSP and DSPP. These features of DCN may contribute to blocking intrafibrillar mineralization in COL I fibrils during the polymer-induced liquid-precursor mineralization process when the fibrils are cross-linked with DCN. This effect was more remarkable when the fibrils were phosphorylated by sodium trimetaphosphate, as shown by the observation of a tube-like morphology via TEM and mineral sheath via SEM. This study enhances the understanding of the role of DCN in mineralization regulation among periodontal tissues. This provides insights for the development of biomaterials for the regeneration of interfaces between soft and hard tissues.
Collapse
Affiliation(s)
- Yuming Bai
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Peng Wu
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Qiufang Zhang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Feng Lin
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Ling Hu
- Department of Pharmacy and Pharmaceutical Sciences, Xiamen Medical College, Xiamen, PR China
| | - Zhisheng Zhang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Wenxia Huang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Yin Xiao
- School of Medicine and Dentistry, Griffith University, Gold Coast, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Qiliang Zuo
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| |
Collapse
|
33
|
Fujie Y, Iwasaki K, Hamasaki M, Suzuki Y, Matsuoka M, Onodera T, Kondo E, Iwasaki N. Paradoxical Change in Subchondral Bone Density in the Medial Compartment of the Proximal Tibial Articular Surface After High Tibial Osteotomy: A Detailed Subchondral Bone Density Analysis. Am J Sports Med 2024; 52:2278-2286. [PMID: 38898822 DOI: 10.1177/03635465241256100] [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] [Indexed: 06/21/2024]
Abstract
BACKGROUND High tibial osteotomy (HTO) aims to realign the varus knee to alleviate stress in the medial compartment. However, detailed information on the impact of HTO on stress distribution across the tibiofemoral joint surface still needs to be completely elucidated. PURPOSE/HYPOTHESIS The present study aimed to analyze the subchondral bone density distribution to validate the alignment threshold causing paradoxical changes. We hypothesized that there would be a paradoxical stress change in the medial compartment beyond a specific threshold for lower limb realignment after HTO. STUDY DESIGN Case series; Level of evidence, 4. METHODS A retrospective clinical study of 32 knees from 30 patients who underwent medial opening-wedge HTO between 2015 and 2019 was conducted at Hokkaido University Hospital. The subchondral bone density across the tibiofemoral joint was analyzed using computed tomography-osteoabsorptiometry before and after HTO. The high-density area (HDA) within the medial and lateral compartments and their subregions, which were quartered in the coronal plane, was specifically examined. RESULTS The hip-knee-ankle angle, medial proximal tibial angle (MPTA), joint line obliquity (JLO), and joint line convergence angle significantly changed after HTO (P < .01). The HDA of the medial compartment to the total HDA ratio decreased from 83% to 77%. Paradoxically, the HDA in the most central subregion of the medial compartment increased from 24% to 30%. There were significant differences between MPTA and JLO in patients with and without paradoxical changes in the HDA. MPTA and JLO cutoff values causing paradoxical changes in the HDA were 94° and 4°, respectively. CONCLUSION There was a paradoxical stress increase in the M4 region at the medial compartment associated with the MPTA and JLO beyond specific thresholds. Therefore, surgical planning should be cautiously performed to prevent overcorrection, which can lead to adverse stress distribution changes.
Collapse
Affiliation(s)
- Yuki Fujie
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Koji Iwasaki
- Department of Functional Reconstruction for the Knee Joint, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanari Hamasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Suzuki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masatake Matsuoka
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Onodera
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Eiji Kondo
- Centre for Sports Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
34
|
Ma Z, Hu X, Li X, An Q, Zhang Y, Guo C, Zhao Y, Zhang Y. Shear Flow-Assembled Janus Membrane with Bifunctional Osteogenic and Antibacterial Effects for Guided Bone Regeneration. ACS Biomater Sci Eng 2024; 10:3984-3993. [PMID: 38728538 DOI: 10.1021/acsbiomaterials.4c00420] [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: 05/12/2024]
Abstract
Guided bone regeneration (GBR) membranes that reside at the interface between the bone and soft tissues for bone repair attract increasing attention, but currently developed GBR membranes suffer from relatively poor osteogenic and antibacterial effects as well as limited mechanical property and biodegradability. We present here the design and fabrication of a bifunctional Janus GBR membrane based on a shear flow-driven layer by a layer self-assembly approach. The Janus GBR membrane comprises a calcium phosphate-collagen/polyethylene glycol (CaP@COL/PEG) layer and a chitosan/poly(acrylic acid) (CHI/PAA) layer on different sides of a collagen membrane to form a sandwich structure. The membrane exhibits good mechanical stability and tailored biodegradability. It is found that the CaP@COL/PEG layer and CHI/PAA layer contribute to the osteogenic differentiation and antibacterial function, respectively. In comparison with the control group, the Janus GBR membrane displays a 2.52-time and 1.84-time enhancement in respective volume and density of newly generated bone. The greatly improved bone repair ability of the Janus GBR membrane is further confirmed through histological analysis, and it has great potential for practical applications in bone tissue engineering.
Collapse
Affiliation(s)
- Zequn Ma
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou 215009, Jiangsu, China
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Xiantong Hu
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing Engineering Research Center of Orthopedics Implants, Beijing 100048, China
| | - Xiangming Li
- Department of Functional Materials, School of Materials Sciences and Technology, Guangdong University of Petrochemical Technology, Maoming 525000, China
| | - Qi An
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| | - Yi Zhang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou 215009, Jiangsu, China
| | - Chunxian Guo
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, 99 Xuefu Road, Suzhou 215009, Jiangsu, China
| | - Yantao Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing Engineering Research Center of Orthopedics Implants, Beijing 100048, China
| | - Yihe Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Sciences and Technology, China University of Geosciences, Beijing 100083, China
| |
Collapse
|
35
|
Liu X, Kong K, Wang J, Liu Z, Tang R. Molecular Weight-Dependent Physiochemical Behaviors of Calcium Carbonate Chains. J Phys Chem Lett 2024; 15:5905-5913. [PMID: 38809103 DOI: 10.1021/acs.jpclett.4c01026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The regulation of physiochemical behaviors by changing molecular weights is an important cornerstone of polymer physics. However, similar correlations between molecular weights and properties have not been discovered in inorganic ionic compounds. In this work, we prepared a calcium carbonate specimen with a semiflexible chain topology analogous to those of polymers. The molecular weights of the calcium carbonate chains, which ranged from 3400 to 54 100 Da, were directly correlated to their physiochemical behaviors, including gel point, zero shear viscosity, and plateau modulus. The calcium carbonate chains showed similar polymeric characteristics, including shear thinning, thixotropy, entropic elasticity, and viscoelasticity. These features agreed with recent theories and formulas in polymer physics textbooks. On the basis of this understanding, the mechanical properties of calcium carbonate-based gels could be altered by changing their molecular weights. This study could represent a fusion of inorganic chemistry and polymer physics with similar molecular weight-dependent behaviors and material properties, establishing an alternative pathway for designing future inorganic materials.
Collapse
Affiliation(s)
- Xin Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Kangren Kong
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Jie Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Zhaoming Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, Zhejiang University, Hangzhou 310027, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
36
|
Bracher S, Voumard B, Simon M, Kochetkova T, Pretterklieber M, Zysset P. Bone collagen tensile properties of the aging human proximal femur. Bone Rep 2024; 21:101773. [PMID: 38778833 PMCID: PMC11109327 DOI: 10.1016/j.bonr.2024.101773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/11/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Despite the dominant role of bone mass in osteoporotic fractures, aging bone tissue properties must be thoroughly understood to improve osteoporosis management. In this context, collagen content and integrity are considered important factors, although limited research has been conducted on the tensile behavior of demineralized compact bone in relation to its porosity and elastic properties in the native mineralized state. Therefore, this study aims (i) at examining the age-dependency of mineralized bone and collagen micromechanical properties; (ii) to test whether, and if so to which extent, collagen properties contribute to mineralized bone mechanical properties. Two cylindrical cortical bone samples from fresh frozen human anatomic donor material were extracted from 80 proximal diaphyseal sections from a cohort of 24 female and 19 male donors (57 to 96 years at death). One sample per section was tested in uniaxial tension under hydrated conditions. First, the native sample was tested elastically (0.25 % strain), and after demineralization, up to failure. Morphology and composition of the second specimen was assessed using micro-computed tomography, Raman spectroscopy, and gravimetric methods. Simple and multiple linear regression were employed to relate morphological, compositional, and mechanical variables with age and sex. Macro-tensile properties revealed that only elastic modulus of native samples was age dependent whereas apparent elastic modulus was sex dependent (p < 0.01). Compositional and morphological analysis detected a weak but significant age and sex dependency of relative mineral weight (r = -0.24, p < 0.05) and collagen disorder ratio (I∼1670/I∼1640, r = 0.25, p < 0.05) and a strong sex dependency of bone volume fraction while generally showing consistent results in mineral content assessment. Young's modulus of demineralized bone was significantly related to tissue mineral density and Young's modulus of native bone. The results indicate that mechanical properties of the organic phase, that include collagen and non-collagenous proteins, are independent of donor age. The observed reduction in relative mineral weight and corresponding overall stiffer response of the collagen network may be caused by a reduced number of mineral-collagen connections and a lack of extrafibrillar and intrafibrillar mineralization that induces a loss of waviness and a collagen fiber pre-stretch.
Collapse
Affiliation(s)
- Stefan Bracher
- ARTORG Center for Biomedical Engineering Research, University of Bern, Switzerland
| | - Benjamin Voumard
- ARTORG Center for Biomedical Engineering Research, University of Bern, Switzerland
| | - Mathieu Simon
- ARTORG Center for Biomedical Engineering Research, University of Bern, Switzerland
| | - Tatiana Kochetkova
- ARTORG Center for Biomedical Engineering Research, University of Bern, Switzerland
| | - Michael Pretterklieber
- Division of Macroscopic and Clinical Anatomy, Gottfried Schatz Research Center, Medical University of Graz, Austria
- Division of Anatomy, Center for Anatomy and Cell Biology, Medical University of Vienna, Austria
| | - Philippe Zysset
- ARTORG Center for Biomedical Engineering Research, University of Bern, Switzerland
| |
Collapse
|
37
|
Tan Y, Jia Z, Deng Z, Li L. Elasmoid fish scales as a natural fibre composite: microscopic heterogeneities in structure, mineral distribution, and mechanical properties. Interface Focus 2024; 14:20230074. [PMID: 39081626 PMCID: PMC11285402 DOI: 10.1098/rsfs.2023.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 08/02/2024] Open
Abstract
The elasmoid scales in teleost fish serve as exemplary models for natural fibre composites with integrated flexibility and protection. Yet, limited research has been focused on the potential structural, chemical, and mechanical heterogeneity within individual scales. This study presents systematic characterizations of the elasmoid scales from black drum fish (Pogonias cromis) at different zones within individual scales as a natural fibre composite, focusing on the microscopic structural heterogeneities and corresponding mechanical effects. The focus field at the centre of the scales exhibits a classical tri-layered collagen-based composite design, consisting of the mineralized outermost limiting layer, external elasmodine layer in the middle, and the unmineralized internal elasmodine layer. In comparison, the rostral field at the anterior end of the scales exhibits a two-layered design: the mineralized outermost limiting layer exhibits radii sections on the outer surface, and the inner elasmodine layer consists of collagen fibre-based sublayers with alternating mineralization levels. Chemical and nanoindentation analysis suggests a close correlation between the mineralization levels and the local nanomechanical properties. Comparative finite element modelling shows that the rostral-field scales achieve increased flexibility under both concave and convex bending. Moreover, the evolving geometries of isolated Mandle's corpuscles in the internal elasmodine layer, transitioning from irregular shapes to faceted octahedrons, suggest the mechanisms of mineral growth and space-filling to thicken the mineralized layers in scales during growth, which enhances the bonding strength between the adjacent collagen fibre layers. This work offers new insights into the structural variations in individual elasmoid scales, providing strategies for bioinspired fibre composite designs with local-adapted functional requirements.
Collapse
Affiliation(s)
- Yiming Tan
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA24061, USA
| | - Zian Jia
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA24061, USA
| | - Zhifei Deng
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA24061, USA
- Hopkins Extreme Materials Institute, Johns Hopkins University, Baltimore, MD21218, USA
| | - Ling Li
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA24061, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA19104, USA
| |
Collapse
|
38
|
Zheng K, Zhong J, Hu J, Nebbiolo E, Sanchez-Weatherby J, Tang T, Landis WJ, Chen J, Winlove P, Sherlock BE, Bell J. Effects of mineralization on the hierarchical organization of collagen-a synchrotron X-ray scattering and polarized second harmonic generation study. Interface Focus 2024; 14:20230046. [PMID: 39081623 PMCID: PMC11285761 DOI: 10.1098/rsfs.2023.0046] [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: 09/30/2023] [Revised: 11/17/2023] [Accepted: 04/22/2024] [Indexed: 08/02/2024] Open
Abstract
The process of mineralization fundamentally alters collagenous tissue biomechanics. While the structure and organization of mineral particles have been widely studied, the impact of mineralization on collagen matrix structure, particularly at the molecular scale, requires further investigation. In this study, synchrotron X-ray scattering (XRD) and polarization-resolved second harmonic generation microscopy (pSHG) were used to study normally mineralizing turkey leg tendon in tissue zones representing different stages of mineralization. XRD data demonstrated statistically significant differences in collagen D-period, intermolecular spacing, fibril and molecular dispersion and relative supramolecular twists between non-mineralizing, early mineralizing and late mineralizing zones. pSHG analysis of the same tendon zones showed the degree of collagen fibril organization was significantly greater in early and late mineralizing zones compared to non-mineralizing zones. The combination of XRD and pSHG data provide new insights into hierarchical collagen-mineral interactions, notably concerning possible cleavage of intra- or interfibrillar bonds, occlusion and reorganization of collagen by mineral with time. The complementary application of XRD and fast, label-free and non-destructive pSHG optical measurements presents a pathway for future investigations into the dynamics of molecular scale changes in collagen in the presence of increasing mineral deposition.
Collapse
Affiliation(s)
- Keke Zheng
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
- Institute for Mechanical Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, University of Sydney, Sydney, Australia
| | - Jingrui Hu
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
| | - Eve Nebbiolo
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
| | | | - Tengteng Tang
- Materials Science & Engineering, McMaster University, Hamilton, Ontario, Canada
| | - William J. Landis
- Preventive and Restorative Dental Sciences, School of Dentistry, University of California at San Francisco, San Francisco, CA, USA
| | - Junning Chen
- Biomedical Engineering, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
| | - Peter Winlove
- Physics and Astronomy, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
| | - Benjamin E. Sherlock
- Physics and Astronomy, Faculty of Environment, Science and Economy, University of Exeter, Exeter, UK
| | - James Bell
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
39
|
Cuahtecontzi Delint R, Jaffery H, Ishak MI, Nobbs AH, Su B, Dalby MJ. Mechanotransducive surfaces for enhanced cell osteogenesis, a review. BIOMATERIALS ADVANCES 2024; 160:213861. [PMID: 38663159 DOI: 10.1016/j.bioadv.2024.213861] [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: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/12/2024] [Indexed: 05/04/2024]
Abstract
Novel strategies employing mechano-transducing materials eliciting biological outcomes have recently emerged for controlling cellular behaviour. Targeted cellular responses are achieved by manipulating physical, chemical, or biochemical modification of material properties. Advances in techniques such as nanopatterning, chemical modification, biochemical molecule embedding, force-tuneable materials, and artificial extracellular matrices are helping understand cellular mechanotransduction. Collectively, these strategies manipulate cellular sensing and regulate signalling cascades including focal adhesions, YAP-TAZ transcription factors, and multiple osteogenic pathways. In this minireview, we are providing a summary of the influence that these materials, particularly titanium-based orthopaedic materials, have on cells. We also highlight recent complementary methodological developments including, but not limited to, the use of metabolomics for identification of active biomolecules that drive cellular differentiation.
Collapse
Affiliation(s)
- Rosalia Cuahtecontzi Delint
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Hussain Jaffery
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mohd I Ishak
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Angela H Nobbs
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Bo Su
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
40
|
Wan Z, Bai X, Wang X, Guo X, Wang X, Zhai M, Fu Y, Liu Y, Zhang P, Zhang X, Yang R, Liu Y, Lv L, Zhou Y. Mgp High-Expressing MSCs Orchestrate the Osteoimmune Microenvironment of Collagen/Nanohydroxyapatite-Mediated Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308986. [PMID: 38588510 PMCID: PMC11187922 DOI: 10.1002/advs.202308986] [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: 11/23/2023] [Revised: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Activating autologous stem cells after the implantation of biomaterials is an important process to initiate bone regeneration. Although several studies have demonstrated the mechanism of biomaterial-mediated bone regeneration, a comprehensive single-cell level transcriptomic map revealing the influence of biomaterials on regulating the temporal and spatial expression patterns of mesenchymal stem cells (MSCs) is still lacking. Herein, the osteoimmune microenvironment is depicted around the classical collagen/nanohydroxyapatite-based bone repair materials via combining analysis of single-cell RNA sequencing and spatial transcriptomics. A group of functional MSCs with high expression of matrix Gla protein (Mgp) is identified, which may serve as a pioneer subpopulation involved in bone repair. Remarkably, these Mgp high-expressing MSCs (MgphiMSCs) exhibit efficient osteogenic differentiation potential and orchestrate the osteoimmune microenvironment around implanted biomaterials, rewiring the polarization and osteoclastic differentiation of macrophages through the Mdk/Lrp1 ligand-receptor pair. The inhibition of Mdk/Lrp1 activates the pro-inflammatory programs of macrophages and osteoclastogenesis. Meanwhile, multiple immune-cell subsets also exhibit close crosstalk between MgphiMSCs via the secreted phosphoprotein 1 (SPP1) signaling pathway. These cellular profiles and interactions characterized in this study can broaden the understanding of the functional MSC subpopulations at the early stage of biomaterial-mediated bone regeneration and provide the basis for materials-designed strategies that target osteoimmune modulation.
Collapse
Affiliation(s)
- Zhuqing Wan
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaoqiang Bai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xin Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiaodong Guo
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xu Wang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Mo Zhai
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yang Fu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yunsong Liu
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ping Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Xiao Zhang
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Ruili Yang
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Yan Liu
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
- Department of OrthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
| | - Longwei Lv
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| | - Yongsheng Zhou
- Department of ProsthodonticsPeking University School and Hospital of StomatologyHaidian DistrictBeijing100081China
- National Center for Stomatology, National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, NHC Key Laboratory of Digital Stomatology, Key Laboratory of Digital StomatologyChinese Academy of Medical SciencesHaidian DistrictBeijing100081China
| |
Collapse
|
41
|
Yu HP, Zhu YJ. Guidelines derived from biomineralized tissues for design and construction of high-performance biomimetic materials: from weak to strong. Chem Soc Rev 2024; 53:4490-4606. [PMID: 38502087 DOI: 10.1039/d2cs00513a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Living organisms in nature have undergone continuous evolution over billions of years, resulting in the formation of high-performance fracture-resistant biomineralized tissues such as bones and teeth to fulfill mechanical and biological functions, despite the fact that most inorganic biominerals that constitute biomineralized tissues are weak and brittle. During the long-period evolution process, nature has evolved a number of highly effective and smart strategies to design chemical compositions and structures of biomineralized tissues to enable superior properties and to adapt to surrounding environments. Most biomineralized tissues have hierarchically ordered structures consisting of very small building blocks on the nanometer scale (nanoparticles, nanofibers or nanoflakes) to reduce the inherent weaknesses and brittleness of corresponding inorganic biominerals, to prevent crack initiation and propagation, and to allow high defect tolerance. The bioinspired principles derived from biomineralized tissues are indispensable for designing and constructing high-performance biomimetic materials. In recent years, a large number of high-performance biomimetic materials have been prepared based on these bioinspired principles with a large volume of literature covering this topic. Therefore, a timely and comprehensive review on this hot topic is highly important and contributes to the future development of this rapidly evolving research field. This review article aims to be comprehensive, authoritative, and critical with wide general interest to the science community, summarizing recent advances in revealing the formation processes, composition, and structures of biomineralized tissues, providing in-depth insights into guidelines derived from biomineralized tissues for the design and construction of high-performance biomimetic materials, and discussing recent progress, current research trends, key problems, future main research directions and challenges, and future perspectives in this exciting and rapidly evolving research field.
Collapse
Affiliation(s)
- Han-Ping Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P. R. China.
| | - Ying-Jie Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P. R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
42
|
Yi Y, An HW, Wang H. Intelligent Biomaterialomics: Molecular Design, Manufacturing, and Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305099. [PMID: 37490938 DOI: 10.1002/adma.202305099] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Materialomics integrates experiment, theory, and computation in a high-throughput manner, and has changed the paradigm for the research and development of new functional materials. Recently, with the rapid development of high-throughput characterization and machine-learning technologies, the establishment of biomaterialomics that tackles complex physiological behaviors has become accessible. Breakthroughs in the clinical translation of nanoparticle-based therapeutics and vaccines have been observed. Herein, recent advances in biomaterials, including polymers, lipid-like materials, and peptides/proteins, discovered through high-throughput screening or machine learning-assisted methods, are summarized. The molecular design of structure-diversified libraries; high-throughput characterization, screening, and preparation; and, their applications in drug delivery and clinical translation are discussed in detail. Furthermore, the prospects and main challenges in future biomaterialomics and high-throughput screening development are highlighted.
Collapse
Affiliation(s)
- Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
43
|
Sauer K, Silveira A, Schoeppler V, Rack A, Zizak I, Pacureanu A, Nassif N, Mantouvalou I, de Nolf W, Fleck C, Shahar R, Zaslansky P. Nanocrystal residual strains and density layers enhance failure resistance in the cleithrum bone of evolutionary advanced pike fish. Acta Biomater 2024; 179:164-179. [PMID: 38513725 DOI: 10.1016/j.actbio.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
Failure-resistant designs are particularly crucial for bones subjected to rapid loading, as is the case for the ambush-hunting northern pike (Esox lucius). These fish have slim and low-density osteocyte-lacking bones. As part of the swallowing mechanism, the cleithrum bone opens and closes the jaw. The cleithrum needs sufficient strength and damage tolerance, to withstand years of repetitive rapid gape-and-suck cycles of feeding. The thin wing-shaped bone comprises anisotropic layers of mineralized collagen fibers that exhibit periodic variations in mineral density on the mm and micrometer length scales. Wavy collagen fibrils interconnect these layers yielding a highly anisotropic structure. Hydrated cleithra exhibit Young's moduli spanning 3-9 GPa where the yield stress of ∼40 MPa increases markedly to exceed ∼180 MPa upon drying. This 5x observation of increased strength corresponds to a change to brittle fracture patterns. It matches the emergence of compressive residual strains of ∼0.15% within the mineral crystals due to forces from shrinking collagen layers. Compressive stresses on the nanoscale, combined with the layered anisotropic microstructure on the mm length scale, jointly confer structural stability in the slender and lightweight bones. By employing a range of X-ray, electron and optical imaging and mechanical characterization techniques, we reveal the structure and properties that make the cleithra impressively damage resistant composites. STATEMENT OF SIGNIFICANCE: By combining structural and mechanical characterization techniques spanning the mm to the sub-nanometer length scales, this work provides insights into the structural organization and properties of a resilient bone found in pike fish. Our observations show how the anosteocytic bone within the pectoral gridle of these fish, lacking any biological (remodeling) repair mechanisms, is adapted to sustain natural repeated loading cycles of abrupt jaw-gaping and swallowing. We find residual strains within the mineral apatite nanocrystals that contribute to forming a remarkably resilient composite material. Such information gleaned from bony structures that are different from the usual bones of mammals showcases how nature incorporates smart features that induce damage tolerance in bone material, an adaptation acquired through natural evolutionary processes.
Collapse
Affiliation(s)
- Katrein Sauer
- Department for Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Aßmannshauser Straße 4-6, 14197 Berlin, Germany.
| | - Andreia Silveira
- Department for Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Aßmannshauser Straße 4-6, 14197 Berlin, Germany
| | - Vanessa Schoeppler
- ESRF- The European Synchrotron, 71 Av. des Martyrs, Grenoble 38000, France
| | - Alexander Rack
- ESRF- The European Synchrotron, 71 Av. des Martyrs, Grenoble 38000, France
| | - Ivo Zizak
- Helmholtz-Zentrum Berlin für Materialien und Energie, Albert-Einstein-Straße 15, Berlin 12489, Germany
| | | | - Nadine Nassif
- CNRS, Sorbonne Université, Collège de FranceLaboratoire Chimie de la Matière Condensée de Paris (LCMCP), Paris F-75005, France
| | - Ioanna Mantouvalou
- Helmholtz-Zentrum Berlin für Materialien und Energie, Albert-Einstein-Straße 15, Berlin 12489, Germany
| | - Wout de Nolf
- ESRF- The European Synchrotron, 71 Av. des Martyrs, Grenoble 38000, France
| | - Claudia Fleck
- Materials Science & Engineering, University of Technology Berlin, Str. des 17. Juni 135 - Sekr. EB 13, Berlin 10623, Germany
| | - Ron Shahar
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Paul Zaslansky
- Department for Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Aßmannshauser Straße 4-6, 14197 Berlin, Germany.
| |
Collapse
|
44
|
Jacobson AM, Zhao X, Sommer S, Sadik F, Warden SJ, Newman C, Siegmund T, Allen MR, Surowiec RK. A comprehensive set of ultrashort echo time magnetic resonance imaging biomarkers to assess cortical bone health: A feasibility study at clinical field strength. Bone 2024; 181:117031. [PMID: 38311304 PMCID: PMC10923147 DOI: 10.1016/j.bone.2024.117031] [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: 11/29/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Conventional bone imaging methods primarily use X-ray techniques to assess bone mineral density (BMD), focusing exclusively on the mineral phase. This approach lacks information about the organic phase and bone water content, resulting in an incomplete evaluation of bone health. Recent research highlights the potential of ultrashort echo time magnetic resonance imaging (UTE MRI) to measure cortical porosity and estimate BMD based on signal intensity. UTE MRI also provides insights into bone water distribution and matrix organization, enabling a comprehensive bone assessment with a single imaging technique. Our study aimed to establish quantifiable UTE MRI-based biomarkers at clinical field strength to estimate BMD and microarchitecture while quantifying bound water content and matrix organization. METHODS Femoral bones from 11 cadaveric specimens (n = 4 males 67-92 yrs of age, n = 7 females 70-95 yrs of age) underwent dual-echo UTE MRI (3.0 T, 0.45 mm resolution) with different echo times and high resolution peripheral quantitative computed tomography (HR-pQCT) imaging (60.7 μm voxel size). Following registration, a 4.5 mm HR-pQCT region of interest was divided into four quadrants and used across the multi-modal images. Statistical analysis involved Pearson correlation between UTE MRI porosity index and a signal-intensity technique used to estimate BMD with corresponding HR-pQCT measures. UTE MRI was used to calculate T1 relaxation time and a novel bound water index (BWI), compared across subregions using repeated measures ANOVA. RESULTS The UTE MRI-derived porosity index and signal-intensity-based estimated BMD correlated with the HR-pQCT variables (porosity: r = 0.73, p = 0.006; BMD: r = 0.79, p = 0.002). However, these correlations varied in strength when we examined each of the four quadrants (subregions, r = 0.11-0.71). T1 relaxometry and the BWI exhibited variations across the four subregions, though these differences were not statistically significant. Notably, we observed a strong negative correlation between T1 relaxation time and the BWI (r = -0.87, p = 0.0006). CONCLUSION UTE MRI shows promise for being an innocuous method for estimating cortical porosity and BMD parameters while also giving insight into bone hydration and matrix organization. This method offers the potential to equip clinicians with a more comprehensive array of imaging biomarkers to assess bone health without the need for invasive or ionizing procedures.
Collapse
Affiliation(s)
- Andrea M Jacobson
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Xuandong Zhao
- Dept. of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Stefan Sommer
- Swiss Center for Musculoskeletal Imaging (SCMI), Balgrist Campus, Zurich, Switzerland; Advanced Clinical Imaging Technology (ACIT), Siemens Healthineers International AG, Zurich, Switzerland.
| | - Farhan Sadik
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Stuart J Warden
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Indianapolis, Indianapolis, IN, USA.
| | - Christopher Newman
- Dept. of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Thomas Siegmund
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Matthew R Allen
- Dept. of Anatomy, Physiology, and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Rachel K Surowiec
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Dept. of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
45
|
Shen D, Zhou Z, Xu Y, Shao C, Shi Y, Zhao W, Tang R, Pan H, Yu M, Hannig M, Fu B. Reversion of ACP Nanoparticles into Prenucleation Clusters via Surfactant for Promoting Biomimetic Mineralization: A Physicochemical Understanding of Biosurfactant Role in Biomineralization Process. Adv Healthc Mater 2024; 13:e2303488. [PMID: 38265149 DOI: 10.1002/adhm.202303488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/21/2023] [Indexed: 01/25/2024]
Abstract
Amphiphilic biomolecules are abundant in mineralization front of biological hard tissues, which play a vital role in osteogenesis and dental hard tissue formation. Amphiphilic biomolecules function as biosurfactants, however, their biosurfactant role in biomineralization process has never been investigated. This study, for the first time, demonstrates that aggregated amorphous calcium phosphate (ACP) nanoparticles can be reversed into dispersed ultrasmall prenucleation clusters (PNCs) via breakdown and dispersion of the ACP nanoparticles by a surfactant. The reduced surface energy of ACP@TPGS and the electrostatic interaction between calcium ions and the pair electrons on oxygen atoms of C-O-C of D-α-tocopheryl polyethylene glycol succinate (TPGS) provide driving force for breakdown and dispersion of ACP nanoparticles into ultrasmall PNCs which promote in vitro and in vivo biomimetic mineralization. The ACP@TPGS possesses excellent biocompatibility without any irritations to oral mucosa and dental pulp. This study not only introduces surfactant into biomimetic mineralization field, but also excites attention to the neglected biosurfactant role during biomineralization process.
Collapse
Affiliation(s)
- Dongni Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Zihuai Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Yuedan Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Changyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Ying Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Weijia Zhao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Ruikang Tang
- Center for Biomaterials and Biopathways, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310000, China
| | - Haihua Pan
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang Province, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, 66424, Homburg, Saarland, Germany
| | - Baiping Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, 310000, China
| |
Collapse
|
46
|
Wang Y, Guan Q, Guo Y, Sun L, Neisiany RE, Guo X, Huang H, Yang L, You Z. Bone-inspired stress-gaining elastomer enabled by dynamic molecular locking. SCIENCE ADVANCES 2024; 10:eadk5177. [PMID: 38517961 PMCID: PMC10959417 DOI: 10.1126/sciadv.adk5177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
The limited capacity of typical materials to resist stress loading, which affects their mechanical performance, is one of the most formidable challenges in materials science. Here, we propose a bone-inspired stress-gaining concept of converting typically destructive stress into a favorable factor to substantially enhance the mechanical properties of elastomers. The concept was realized by a molecular design of dynamic poly(oxime-urethanes) network with mesophase domains. During external loading, the mesophase domains in the condensed state were aligned into more ordered domains, and the dynamic oxime-urethane bonds served as the dynamic molecular locks disassociating and reorganizing to facilitate and fix the mesophase domains. Consequently, the tensile modulus and strength were enhanced by 1744 and 49.3 times after four cycles of mechanical training, respectively. This study creates a molecular concept with stress-gaining properties induced by repeated mechanical stress loading and will inspire a series of innovative materials for diverse applications.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Qingbao Guan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Yue Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Lijie Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Rasoul Esmaeely Neisiany
- Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| | - Xuran Guo
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongfei Huang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Lei Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| |
Collapse
|
47
|
Zhao Y, Sun W, Wu X, Gao X, Song F, Duan B, Lu A, Yang H, Huang C. Janus Membrane with Intrafibrillarly Strontium-Apatite-Mineralized Collagen for Guided Bone Regeneration. ACS NANO 2024; 18:7204-7222. [PMID: 38373291 DOI: 10.1021/acsnano.3c12403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Commercial collagen membranes face difficulty in guided bone regeneration (GBR) due to the absence of hierarchical structural design, effective interface management, and diverse bioactivity. Herein, a Janus membrane called SrJM is developed that consists of a porous collagen face to enhance osteogenic function and a dense face to maintain barrier function. Specifically, biomimetic intrafibrillar mineralization of collagen with strontium apatite is realized by liquid precursors of amorphous strontium phosphate. Polycaprolactone methacryloyl is further integrated on one side of the collagen as a dense face, which endows SrJM with mechanical support and a prolonged lifespan. In vitro experiments demonstrate that the dense face of SrJM acts as a strong barrier against fibroblasts, while the porous face significantly promotes cell adhesion and osteogenic differentiation through activation of calcium-sensitive receptor/integrin/Wnt signaling pathways. Meanwhile, SrJM effectively enhances osteogenesis and angiogenesis by recruiting stem cells and modulating osteoimmune response, thus creating an ideal microenvironment for bone regeneration. In vivo studies verify that the bone defect region guided by SrJM is completely repaired by newly formed vascularized bone. Overall, the outstanding performance of SrJM supports its ongoing development as a multifunctional GBR membrane, and this study provides a versatile strategy of fabricating collagen-based biomaterials for hard tissue regeneration.
Collapse
Affiliation(s)
- Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Wei Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Xin Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Bo Duan
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Ang Lu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| |
Collapse
|
48
|
Zhang Y, Wang Y, Zhang Z, Wang Z, Shao C, Hannig M, Zhou Z, Fu B. Intrafibrillar mineralization of type I collagen with calcium carbonate and strontium carbonate induced by polyelectrolyte-cation complexes. NANOSCALE ADVANCES 2024; 6:467-480. [PMID: 38235102 PMCID: PMC10791124 DOI: 10.1039/d3na00705g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024]
Abstract
Calcium carbonate (CaCO3), possessing excellent biocompatibility, bioactivity, osteoconductivity and superior biodegradability, may serve as an alternative to hydroxyapatite (HAp), the natural inorganic component of bone and dentin. Intrafibrillar mineralization of collagen with CaCO3 was achieved through the polymer-induced liquid precursor (PILP) process for at least 2 days. This study aims to propose a novel pathway for rapid intrafibrillar mineralization with CaCO3 by sequential application of the carbonate-bicarbonate buffer and polyaspartic acid (pAsp)-Ca suspension. Fourier transform infrared (FTIR) spectroscopy, zeta potential measurements, atomic force microscopy/Kelvin probe force microscopy (AFM/KPFM), and three-dimensional stochastic optical reconstruction microscopy (3D STORM) demonstrated that the carbonate-bicarbonate buffer significantly decreased the surface potential of collagen and CO32-/HCO3- ions could attach to collagen fibrils via hydrogen bonds. The electropositive pAsp-Ca complexes and free Ca2+ ions are attracted to and interact with CO32-/HCO3- ions through electrostatic attractions to form amorphous calcium carbonate that crystallizes gradually. Moreover, like CaCO3, strontium carbonate (SrCO3) can deposit inside the collagen fibrils through this pathway. The CaCO3-mineralized collagen gels exhibited better biocompatibility and cell proliferation ability than SrCO3. This study provides a feasible strategy for rapid collagen mineralization with CaCO3 and SrCO3, as well as elucidating the tissue engineering of CaCO3-based biomineralized materials.
Collapse
Affiliation(s)
- Yizhou Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Yiru Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Zhengyi Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Zhe Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Changyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University 66421 Homburg Germany
| | - Zihuai Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| | - Baiping Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000
| |
Collapse
|
49
|
Tang K, Xue J, Zhu Y, Wu C. Design and synthesis of bioinspired nanomaterials for biomedical application. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1914. [PMID: 37394619 DOI: 10.1002/wnan.1914] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023]
Abstract
Natural materials and bioprocesses provide abundant inspirations for the design and synthesis of high-performance nanomaterials. In the past several decades, bioinspired nanomaterials have shown great potential in the application of biomedical fields, such as tissue engineering, drug delivery, and cancer therapy, and so on. In this review, three types of bioinspired strategies for biomedical nanomaterials, that is, inspired by the natural structures, biomolecules, and bioprocesses, are mainly introduced. We summarize and discuss the design concepts and synthesis approaches of various bioinspired nanomaterials along with their specific roles in biomedical applications. Additionally, we discuss the challenges for the development of bioinspired biomedical nanomaterials, such as mechanical failure in wet environment, limitation in scale-up fabrication, and lack of deep understanding of biological properties. It is expected that the development and clinical translation of bioinspired biomedical nanomaterials will be further promoted under the cooperation of interdisciplinary subjects in future. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Kai Tang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jianmin Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Zhang Y, Ma S, Nie J, Liu Z, Chen F, Li A, Pei D. Journey of Mineral Precursors in Bone Mineralization: Evolution and Inspiration for Biomimetic Design. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2207951. [PMID: 37621037 DOI: 10.1002/smll.202207951] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/27/2023] [Indexed: 08/26/2023]
Abstract
Bone mineralization is a ubiquitous process among vertebrates that involves a dynamic physical/chemical interplay between the organic and inorganic components of bone tissues. It is now well documented that carbonated apatite, an inorganic component of bone, is proceeded through transient amorphous mineral precursors that transforms into the crystalline mineral phase. Here, the evolution on mineral precursors from their sources to the terminus in the bone mineralization process is reviewed. How organisms tightly control each step of mineralization to drive the formation, stabilization, and phase transformation of amorphous mineral precursors in the right place, at the right time, and rate are highlighted. The paradigm shifts in biomineralization and biomaterial design strategies are intertwined, which promotes breakthroughs in biomineralization-inspired material. The design principles and implementation methods of mineral precursor-based biomaterials in bone graft materials such as implant coatings, bone cements, hydrogels, and nanoparticles are detailed in the present manuscript. The biologically controlled mineralization mechanisms will hold promise for overcoming the barriers to the application of biomineralization-inspired biomaterials.
Collapse
Affiliation(s)
- Yuchen Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shaoyang Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiaming Nie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Faming Chen
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|