1
|
Hung GY, Wang CY, Feng KC, Tu CS, Cheng IC, Mana-Ay H, Hsiao HY, Lai PL, Chen PY. Manipulating Mg/Ca ratios in MgO-CaO-SiO 2 bioactive glass for achieving accelerated osteogenic differentiation of human adipose-derived stem cells. BIOMATERIALS ADVANCES 2025; 169:214189. [PMID: 39826260 DOI: 10.1016/j.bioadv.2025.214189] [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: 10/02/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Cell-containing biomaterial is a promising material for treating nonunion or critical bone defect. Human adipose-derived stem cells (hADSCs) are suitable for bone repair due to their abundance in the abdomen, thighs, and buttocks. However, the low osteogenic capacities of hADSCs hinder their extended development for bone regeneration application. The present goal explores a novel MgO-CaO-SiO2 bioactive glass with suitable Mg/Ca ratios to enhance the osteogenic differentiation and bioactivity of hADSCs. The synthetic bioglass can be expressed as xMgO-(2-x)CaO-SiO2 (abbreviated as Mg(x)Ca(2-x)Si2, x = 0, 0.25, 0.5, 0.75, and 1). The expression levels of osteoblast-related genes (i.e., BMP2, RUNX2, DLX5, COL1A1, BGLAP2, and SPP1) were evaluated by reverse transcription-quantitative PCR (RT-PCR). The proteins involved in the p38/Akt/ERK signaling pathways were analyzed with Western blots. The results indicated that the extractions from the Mg(x)Ca(2-x)Si2 bioglass promoted hADSCs proliferation. Among the Mg(x)Ca(2-x)Si2 bioglass with different Mg/Ca ratios, the bioglass with a low Mg/Ca ratio (x = 0.25) presented greater osteogenic differentiation of hADSCs by promoting the p38 signaling pathway. Interestingly, the bioglass with low Mg/Ca ratio (x = 0.25) further presented on osteogenic potential with greater osteointegration in rat femoral defect model. This work provides the optimal Mg/Ca ratio in Mg(x)Ca(2-x)Si2 bioglass to promote the osteogenic induction of hADSCs and bone regeneration.
Collapse
Affiliation(s)
- Guan-Yi Hung
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Yun Wang
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Kuei-Chih Feng
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Shun Tu
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - I-Chien Cheng
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Haidee Mana-Ay
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Silliman University, Dumaguete City 6200, Philippines
| | - Hui-Yi Hsiao
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Biomedical Sciences, Chang Gung University, Taoyuan City 33305, Taiwan; Center for Tissue Engineering, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan.
| | - Po-Liang Lai
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan.
| | - Pin-Yi Chen
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| |
Collapse
|
2
|
Liang W, Long H, Zhang H, Bai J, Jiang B, Wang J, Fu L, Ming W, Zhao J, Zeng B. Bone scaffolds-based localized drugs delivery for osteosarcoma: current status and future perspective. Drug Deliv 2024; 31:2391001. [PMID: 39239763 PMCID: PMC11382735 DOI: 10.1080/10717544.2024.2391001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
A common malignant bone neoplasm in teenagers is Osteosarcoma. Chemotherapy, surgical therapy, and radiation therapy together comprise the usual clinical course of treatment for Osteosarcoma. While Osteosarcoma and other bone tumors are typically treated surgically, however, surgical resection frequently fails to completely eradicate tumors, and in turn becomes the primary reason for postoperative recurrence and metastasis, ultimately leading to a high rate of mortality. Patients still require radiation and/or chemotherapy after surgery to stop the spread of the tumor and its metastases, and both treatments have an adverse influence on the body's organ systems. In the postoperative management of osteosarcoma, bone scaffolds can load cargos (growth factors or drugs) and function as drug delivery systems (DDSs). This review describes the different kinds of bone scaffolds that are currently available and highlights key studies that use scaffolds as DDSs for the treatment of osteosarcomas. The discussion also includes difficulties and perspectives regarding the use of scaffold-based DDSs. The study may serve as a source for outlining efficient and secure postoperative osteosarcoma treatment plans.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hongwei Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Juqin Bai
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiangwei Wang
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lifeng Fu
- Department of Orthopedics, Shaoxing City Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Wenyi Ming
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bin Zeng
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
3
|
Al-Naymi HAS, Al-Musawi MH, Mirhaj M, Valizadeh H, Momeni A, Danesh Pajooh AM, Shahriari-Khalaji M, Sharifianjazi F, Tavamaishvili K, Kazemi N, Salehi S, Arefpour A, Tavakoli M. Exploring nanobioceramics in wound healing as effective and economical alternatives. Heliyon 2024; 10:e38497. [PMID: 39391491 PMCID: PMC11466581 DOI: 10.1016/j.heliyon.2024.e38497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Wound healing is a sophisticated process for which various treatment methods have been developed. Bioceramics with the ability to release inorganic ions in biological environments play a crucial role in cellular metabolism and exhibit bactericidal activity, contributing to numerous physiological processes. Their multifaceted roles in biological systems highlight their significance. The release of different metallic ions from bioceramics enables the repair of both hard and soft tissues. These ions may be effective in cell motility, proliferation, differentiation, adhesion, angiogenesis, and antibiosis. Unlike conventional medications, the bioactivity and antibacterial properties of bioceramics are typically not associated with side effects or bacterial resistance. Bioceramics are commonly recognized for their capcity to facilitate the healing of hard tissues due to their exceptional mechanical properties. In this review, we first explore wound treatment and its prevalent methods, and subsequently, we discuss the application of three primary categories of bioceramics-oxide ceramics, silicate-based ceramics, and calcium-phosphate ceramics-in the context of wound treatment. This review introduces bioceramics as a cost-effective and efficient alternative for wound repair. Our aim is to inspire researchers to incorporate bioceramics with other biomaterials to achieve enhanced, economical, expedited, and safer wound healing.
Collapse
Affiliation(s)
- Hanan Adnan Shaker Al-Naymi
- Department of Chemistry, College of Education for Pure Science/Ibn Al-Haitham, University of Baghdad, Baghdad, Iraq
| | - Mastafa H. Al-Musawi
- Department of Clinical Laboratory Science, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq
| | - Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| | - Hamideh Valizadeh
- Department of tissue engineering and regenerative medicine, Faculty of advanced technologies in medicine, Iran university of medical sciences, Tehran, Iran
| | - Arefeh Momeni
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Amir Mohammad Danesh Pajooh
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mina Shahriari-Khalaji
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Fariborz Sharifianjazi
- Center for Advanced Materials and Structures, School of Science and Technology, The University of Georgia, 0171, Tbilisi, Georgia
- Department of Civil Engineering, School of Science and Technology, The University of Georgia, 0171, Tbilisi, Georgia
| | - Ketevan Tavamaishvili
- Georgian American University, School of Medicine, 10 Merab Aleksidze Str., Tbilisi, 0160, Georgia
| | - Nafise Kazemi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Saeideh Salehi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Ahmadreza Arefpour
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran
| |
Collapse
|
4
|
Raza A, Wu W. Metal-organic frameworks in oral drug delivery. Asian J Pharm Sci 2024; 19:100951. [PMID: 39493807 PMCID: PMC11530798 DOI: 10.1016/j.ajps.2024.100951] [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: 12/31/2023] [Revised: 05/02/2024] [Accepted: 06/23/2024] [Indexed: 11/05/2024] Open
Abstract
Metal-organic frameworks (MOFs) offer innovative solutions to the limitations of traditional oral drug delivery systems through their unique combination of metal ions and organic ligands. This review systematically examines the structural properties and principles of MOFs, setting the stage for their application in drug delivery. It discusses various classes of MOFs, including those based on zirconium, iron, zinc, copper, titanium, aluminum, potassium, and magnesium, assessing their drug-loading capacities, biocompatibility, and controlled release mechanisms. The effectiveness of MOFs is illustrated through case studies that highlight their capabilities in enhancing drug solubility, providing protection against the harsh gastrointestinal environment, and enabling precise drug release. The review addresses potential challenges, particularly the toxicity concerns associated with MOFs, and calls for further research into their biocompatibility and interactions with biological systems. It concludes by emphasizing the potential of MOFs in revolutionizing oral drug delivery, highlighting the critical need for comprehensive research to harness their full potential in clinical applications.
Collapse
Affiliation(s)
- Aun Raza
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
| |
Collapse
|
5
|
Min KH, Kim DH, Kim KH, Seo JH, Pack SP. Biomimetic Scaffolds of Calcium-Based Materials for Bone Regeneration. Biomimetics (Basel) 2024; 9:511. [PMID: 39329533 PMCID: PMC11430767 DOI: 10.3390/biomimetics9090511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Calcium-based materials, such as calcium carbonate, calcium phosphate, and calcium silicate, have attracted significant attention in biomedical research, owing to their unique physicochemical properties and versatile applications. The distinctive characteristics of these materials, including their inherent biocompatibility and tunable structures, hold significant promise for applications in bone regeneration and tissue engineering. This review explores the biomedical applications of calcium-containing materials, particularly for bone regeneration. Their remarkable biocompatibility, tunable nanostructures, and multifaceted functionalities make them pivotal for advancing regenerative medicine, drug delivery system, and biomimetic scaffold applications. The evolving landscape of biomedical research continues to uncover new possibilities, positioning calcium-based materials as key contributors to the next generation of innovative biomaterial scaffolds.
Collapse
Affiliation(s)
- Ki Ha Min
- Institute of Industrial Technology, Korea University, Sejong 30019, Republic of Korea;
| | - Dong Hyun Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.H.K.); (K.H.K.); (J.-H.S.)
| | - Koung Hee Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.H.K.); (K.H.K.); (J.-H.S.)
| | - Joo-Hyung Seo
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.H.K.); (K.H.K.); (J.-H.S.)
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea; (D.H.K.); (K.H.K.); (J.-H.S.)
| |
Collapse
|
6
|
Li N, Wang J, Feng G, Liu Y, Shi Y, Wang Y, Chen L. Advances in biomaterials for oral-maxillofacial bone regeneration: spotlight on periodontal and alveolar bone strategies. Regen Biomater 2024; 11:rbae078. [PMID: 39055303 PMCID: PMC11272181 DOI: 10.1093/rb/rbae078] [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: 03/03/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/27/2024] Open
Abstract
The intricate nature of oral-maxillofacial structure and function, coupled with the dynamic oral bacterial environment, presents formidable obstacles in addressing the repair and regeneration of oral-maxillofacial bone defects. Numerous characteristics should be noticed in oral-maxillofacial bone repair, such as irregular morphology of bone defects, homeostasis between hosts and microorganisms in the oral cavity and complex periodontal structures that facilitate epithelial ingrowth. Therefore, oral-maxillofacial bone repair necessitates restoration materials that adhere to stringent and specific demands. This review starts with exploring these particular requirements by introducing the particular characteristics of oral-maxillofacial bones and then summarizes the classifications of current bone repair materials in respect of composition and structure. Additionally, we discuss the modifications in current bone repair materials including improving mechanical properties, optimizing surface topography and pore structure and adding bioactive components such as elements, compounds, cells and their derivatives. Ultimately, we organize a range of potential optimization strategies and future perspectives for enhancing oral-maxillofacial bone repair materials, including physical environment manipulation, oral microbial homeostasis modulation, osteo-immune regulation, smart stimuli-responsive strategies and multifaceted approach for poly-pathic treatment, in the hope of providing some insights for researchers in this field. In summary, this review analyzes the complex demands of oral-maxillofacial bone repair, especially for periodontal and alveolar bone, concludes multifaceted strategies for corresponding biomaterials and aims to inspire future research in the pursuit of more effective treatment outcomes.
Collapse
Affiliation(s)
- Nayun Li
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinyu Wang
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guangxia Feng
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuqing Liu
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yunsong Shi
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yifan Wang
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lili Chen
- 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, Union Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Oral and Maxillofacial Medical Devices and Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Kämmerer PW, Heimes D, Zaage F, Ganz C, Frerich B, Gerber T, Dau M. Improving material properties of a poloxamer P407 hydrogel-based hydroxyapatite bone substitute material by adding silica-A comparative in vivo study. J Biomed Mater Res B Appl Biomater 2024; 112:e35405. [PMID: 38701384 DOI: 10.1002/jbm.b.35405] [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: 10/26/2023] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
The structure and handling properties of a P407 hydrogel-based bone substitute material (BSM) might be affected by different poloxamer P407 and silicon dioxide (SiO2) concentrations. The study aimed to compare the mechanical properties and biological parameters (bone remodeling, BSM degradation) of a hydroxyapatite: silica (HA)-based BSM with various P407 hydrogels in vitro and in an in vivo rat model. Rheological analyses for mechanical properties were performed on one BSM with an SiO2-enriched hydrogel (SPH25) as well on two BSMs with unaltered hydrogels in different gel concentrations (PH25 and PH30). Furthermore, the solubility of all BSMs were tested. In addition, 30 male Wistar rats underwent surgical creation of a well-defined bone defect in the tibia. Defects were filled randomly with PH30 (n = 15) or SPH25 (n = 15). Animals were sacrificed after 12 (n = 5 each), 21 (n = 5 each), and 63 days (n = 5 each). Histological evaluation and histomorphometrical quantification of new bone formation (NB;%), residual BSM (rBSM;%), and soft tissue (ST;%) was conducted. Rheological tests showed an increased viscosity and lower solubility of SPH when compared with the other hydrogels. Histomorphometric analyses in cancellous bone showed a decrease of ST in PH30 (p = .003) and an increase of NB (PH30: p = .001; SPH: p = .014) over time. A comparison of both BSMs revealed no significant differences. The addition of SiO2 to a P407 hydrogel-based hydroxyapatite BSM improves its mechanical stability (viscosity, solubility) while showing similar in vivo healing properties compared to PH30. Additionally, the SiO2-enrichment allows a reduction of poloxamer ratio in the hydrogel without impairing the material properties.
Collapse
Affiliation(s)
- Peer W Kämmerer
- Department of Oral, Maxillofacial Plastic Surgery, University Medical Center Mainz, Mainz, Germany
- Department of Oral, Maxillofacial Plastic Surgery, University Medical Center Rostock, Rostock, Germany
| | - Diana Heimes
- Department of Oral, Maxillofacial Plastic Surgery, University Medical Center Mainz, Mainz, Germany
| | | | - Cornelia Ganz
- Institute of Physics, Rostock University, Rostock, Germany
| | - Bernhard Frerich
- Department of Oral, Maxillofacial Plastic Surgery, University Medical Center Rostock, Rostock, Germany
| | - Thomas Gerber
- Institute of Physics, Rostock University, Rostock, Germany
| | - Michael Dau
- Department of Oral, Maxillofacial Plastic Surgery, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
8
|
Mamidi N, Ijadi F, Norahan MH. Leveraging the Recent Advancements in GelMA Scaffolds for Bone Tissue Engineering: An Assessment of Challenges and Opportunities. Biomacromolecules 2024; 25:2075-2113. [PMID: 37406611 DOI: 10.1021/acs.biomac.3c00279] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The field of bone tissue engineering has seen significant advancements in recent years. Each year, over two million bone transplants are performed globally, and conventional treatments, such as bone grafts and metallic implants, have their limitations. Tissue engineering offers a new level of treatment, allowing for the creation of living tissue within a biomaterial framework. Recent advances in biomaterials have provided innovative approaches to rebuilding bone tissue function after damage. Among them, gelatin methacryloyl (GelMA) hydrogel is emerging as a promising biomaterial for supporting cell proliferation and tissue regeneration, and GelMA has exhibited exceptional physicochemical and biological properties, making it a viable option for clinical translation. Various methods and classes of additives have been used in the application of GelMA for bone regeneration, with the incorporation of nanofillers or other polymers enhancing its resilience and functional performance. Despite promising results, the fabrication of complex structures that mimic the bone architecture and the provision of balanced physical properties for both cell and vasculature growth and proper stiffness for load bearing remain as challenges. In terms of utilizing osteogenic additives, the priority should be on versatile components that promote angiogenesis and osteogenesis while reinforcing the structure for bone tissue engineering applications. This review focuses on recent efforts and advantages of GelMA-based composite biomaterials for bone tissue engineering, covering the literature from the last five years.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Fatemeh Ijadi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| |
Collapse
|
9
|
Sun W, Yang K, Zou Y, Ren Y, Zhang L, Zhang F, Zeng R. In vitro and in vivo degradation, biocompatibility and bone repair performance of strontium-doped montmorillonite coating on Mg-Ca alloy. Regen Biomater 2024; 11:rbae027. [PMID: 38605854 PMCID: PMC11007119 DOI: 10.1093/rb/rbae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/22/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024] Open
Abstract
Poor bone growth remains a challenge for degradable bone implants. Montmorillonite and strontium were selected as the carrier and bone growth promoting elements to prepare strontium-doped montmorillonite coating on Mg-Ca alloy. The surface morphology and composition were characterized by SEM, EDS, XPS, FT-IR and XRD. The hydrogen evolution experiment and electrochemical test results showed that the Mg-Ca alloy coated with Sr-MMT coating possessed optimal corrosion resistance performance. Furthermore, in vitro studies on cell activity, ALP activity, and cell morphology confirmed that Sr-MMT coating had satisfactory biocompatibility, which can significantly avail the proliferation, differentiation, and adhesion of osteoblasts. Moreover, the results of the 90-day implantation experiment in rats indicated that, the preparation of Sr-MMT coating effectively advanced the biocompatibility and bone repair performance of Mg-Ca alloy. In addition, The Osteogenic ability of Sr-MMT coating may be due to the combined effect of the precipitation of Si4+ and Sr2+ in Sr-MMT coating and the dissolution of Mg2+ and Ca2+ during the degradation of Mg-Ca alloy. By using coating technology, this study provides a late-model strategy for biodegradable Mg alloys with good corrosion resistance, biocompatibility. This new material will bring more possibilities in bone repair.
Collapse
Affiliation(s)
- Wenxin Sun
- Department of Bioengineering, College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Kaining Yang
- Department of Bioengineering, College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Yuhong Zou
- Department of Bioengineering, College of Chemical and Biological Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Yande Ren
- Affiliated Hospital of Medical College Qingdao University, Qingdao 266555, China
| | - Lin Zhang
- Hospital of Shandong, University of Science and Technology, Qingdao 266590, China
| | - Fen Zhang
- Corrosion Laboratory for Light Metals, College of Materials Science and Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Rongchang Zeng
- Corrosion Laboratory for Light Metals, College of Materials Science and Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| |
Collapse
|
10
|
Zhao Q, Ni Y, Wei H, Duan Y, Chen J, Xiao Q, Gao J, Yu Y, Cui Y, Ouyang S, Miron RJ, Zhang Y, Wu C. Ion incorporation into bone grafting materials. Periodontol 2000 2024; 94:213-230. [PMID: 37823468 DOI: 10.1111/prd.12533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
The use of biomaterials in regenerative medicine has expanded to treat various disorders caused by trauma or disease in orthopedics and dentistry. However, the treatment of large and complex bone defects presents a challenge, leading to a pressing need for optimized biomaterials for bone repair. Recent advances in chemical sciences have enabled the incorporation of therapeutic ions into bone grafts to enhance their performance. These ions, such as strontium (for bone regeneration/osteoporosis), copper (for angiogenesis), boron (for bone growth), iron (for chemotaxis), cobalt (for B12 synthesis), lithium (for osteogenesis/cementogenesis), silver (for antibacterial resistance), and magnesium (for bone and cartilage regeneration), among others (e.g., zinc, sodium, and silica), have been studied extensively. This review aims to provide a comprehensive overview of current knowledge and recent developments in ion incorporation into biomaterials for bone and periodontal tissue repair. It also discusses recently developed biomaterials from a basic design and clinical application perspective. Additionally, the review highlights the importance of precise ion introduction into biomaterials to address existing limitations and challenges in combination therapies. Future prospects and opportunities for the development and optimization of biomaterials for bone tissue engineering are emphasized.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Hongjiang Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiling Duan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jingqiu Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Qi Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Jie 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, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yiqian Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yu Cui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Simin Ouyang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- School of Medicine, Medical Research Institute, Wuhan University, Wuhan, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
11
|
Bahadorani F, Hadadzadeh H, Mirahmadi-Zare SZ, Masaeli E. Nanocore-Shell Bone Filler Contained Mesoporous Silica Modified with Hydroxyapatite Precursors; Wrapped in a Natural Metal-Phenolic Network. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:16090-16100. [PMID: 37921536 DOI: 10.1021/acs.langmuir.3c02227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Various therapeutic strategies have been developed to address bone diseases caused by aging society and skeletal defects caused by trauma or accidental events. One such approach is using bone fillers, such as hydroxyapatite (HA) and bioactive glasses. Although they have provided effective osteogenesis, infection and inflammation due to the surgical procedure and uncontrolled ion release can hinder the efficiency of bone regeneration. In response to these challenges, immobilizing a neutral metal-phenolic network on the surface of osteoconductive nanoparticles would be the master key to achieving a gradual, controlled release during the mineralization period and reducing infection and inflammation through biological pathways. In this regard, a mesoporous silica nanocomposite modified by an HA precursor was synthesized to enhance bone regeneration. In addition, to improve the therapeutic effects, its surface was wrapped with a magnesium-phenolic network made from pomegranate extract, which can simultaneously produce anti-inflammatory and antibacterial effects. The obtained core-shell nanocomposite was characterized by its physicochemical properties, biocompatibility, and bioactivity. The in vitro studies revealed that the synthesized nanocomposite exhibits higher osteogenic activity than the control groups, as confirmed by alizarin red staining. Moreover, the nanocomposite maintained low toxicity as measured by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and increased antibacterial activity against Staphylococcus aureus and Escherichia coli compared with the control groups. Therefore, this research presents a promising strategy for bone regeneration, combining the advantages of mesoporous silica nanocomposite modified by an HA precursor with the beneficial effects of a magnesium-phenolic network.
Collapse
Affiliation(s)
- Fatemeh Bahadorani
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Hassan Hadadzadeh
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Elahe Masaeli
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| |
Collapse
|
12
|
Yang G, Fan R, Yang J, Yi L, Chen S, Wan W. Magnesium/gallic acid bioMOFs laden carbonized mushroom aerogel effectively heals biofilm-infected skin wounds. Biomaterials 2023; 302:122347. [PMID: 37827053 DOI: 10.1016/j.biomaterials.2023.122347] [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: 07/22/2023] [Revised: 09/19/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
Biofilm-infected acute skin wounds are still one of the significant challenges that need to be solved urgently in wound healing. Herein, we reported a magnesium/gallic acid bio-MOFs laden carbonized mushroom aerogel (QMOFs-PCMA) combined with photothermal therapy for eradicating biofilms in skin wounds. The design of bioMOFs is mainly responsible for regulating immunity. In vitro, it exhibited ROS clearance and antioxidant ability. In vivo, it could regulate local immune responses from pro-inflammatory status to pro-regenerative status, resulting in decreased inflammatory cytokines expression and increased anti-inflammatory cytokines expression. The carbonized mushroom aerogel is mainly responsible for photothermal therapy (PTT), and the polydopamine and bioMOFs could enhance the photothermal conversion efficiency and stability of carbonized aerogels. The carbonized aerogel in combination with PTT could eradicate S. aureus biofilm in both in vitro and in vivo studies and clear E. coli biofilms in vitro studies. The biofilm clearance and improved inflammatory responses laid a good foundation for wound healing, resulting in the granulation tissue formation, re-epithelialization, and angiogenesis significantly enhanced in the QMOFs-PCMA + NIR group. Our results indicate that the QMOFs-PCMA combined with photothermal therapy may provide a promising treatment for biofilm-infected skin wounds.
Collapse
Affiliation(s)
- Ganghua Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Ruyi Fan
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jianqiu Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Lei Yi
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China.
| | - Wenbing Wan
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
13
|
Rittidach T, Sillapaprayoon S, Chantho V, Pimtong W, Charoenphandhu N, Thongbunchoo J, Krishnamra N, Bootchanont A, Porjai P, Pon-On W. Investigation on the physical properties and biocompatibility of zirconia-alumina-silicate@diopside composite materials and its in vivo toxicity study in embryonic zebrafish. RSC Adv 2023; 13:30575-30585. [PMID: 37859778 PMCID: PMC10583262 DOI: 10.1039/d3ra04555b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
Bioceramic materials have a wide range of applications in the biomedical field, such as in the repair of bone defects and dental surgery. Silicate-based bioceramics have attracted biomedical researchers' interest due to their bioactivity and biodegradability. In this study, extended the scope of ZAS utilization in bone tissue engineering by introducing calcium-magnesium-silicate (diopside, CMS) as an interface material aim to develop a machinable bioceramic composite (ZASCMS) by the sol-gel method. The physicochemical characterization, in vitro biological properties and in vivo zebrafish cytotoxicity study of ZAS-based composites as a function of CMS contents, 0, 25, 50, 75 and 100 wt%, were performed. Results showed that the as-prepared ZASCMS possessed porous architecture with well-interconnected pore structure. Results also revealed that the mechanical properties of ZASCMS composite materials were gradually improved with increasing CMS contents. The ZASCMS composites with more than 50 wt% CMS had the highest compressive strength and modulus of 6.78 ± 0.62 MPa and 340.10 ± 16.81 MPa, respectively. Regarding in vitro bioactivities, the composite scaffolds were found to stimulate osteoblast-like UMR-106 cell adhesion, growth, and proliferation. The antibacterial activity of the ZASCMS composite scaffolds was tested against Staphylococcus epidermidis (S. epidermidis) and Escherichia coli (E. coli) also exhibited an antibacterial property. Furthermore, the in vivo studies using embryonic zebrafish were exposed to as-prepared particles (0-500 μg mL-1) and showed that the synthesized ZAS, CMS and ZASCMS composite particles were non-toxic based on the evaluation of survivability, hatching rate and embryonic morphology. In conclusions, our results indicated that the synthesized composite exhibited their biological properties and antibacterial activity, which could well be a promising material with high potential to be applied in orthopaedic and dental tissue engineering.
Collapse
Affiliation(s)
- Tanawut Rittidach
- Department of Physics, Faculty of Science, Kasetsart University Bangkok 10900 Thailand
| | - Siwapech Sillapaprayoon
- Nano Environmental and Health Safety Research Team, National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA) Pathum Thani 12120 Thailand
| | - Varissara Chantho
- Nano Environmental and Health Safety Research Team, National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA) Pathum Thani 12120 Thailand
| | - Wittaya Pimtong
- Nano Environmental and Health Safety Research Team, National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA) Pathum Thani 12120 Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Department of Physiology, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Institute of Molecular Biosciences, Mahidol University Nakhon Pathom 73170 Thailand
- The Academy of Science, The Royal Society of Thailand Dusit Bangkok 10300 Thailand
| | - Jirawan Thongbunchoo
- Center of Calcium and Bone Research, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Department of Physiology, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Nateetip Krishnamra
- Center of Calcium and Bone Research, Faculty of Science, Mahidol University Bangkok 10400 Thailand
- Department of Physiology, Faculty of Science, Mahidol University Bangkok 10400 Thailand
| | - Atipong Bootchanont
- Division of Physics, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi Pathum Thani 12120 Thailand
| | - Porramain Porjai
- Division of Physics, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi Pathum Thani 12120 Thailand
| | - Weeraphat Pon-On
- Department of Physics, Faculty of Science, Kasetsart University Bangkok 10900 Thailand
| |
Collapse
|
14
|
Vijayakumar N, Venkatraman SK, Nandakumar R, Alex RA, Abraham J, Mohammadi H, Ebadi M, Swamiappan S. Optimization of Metal Ion/Fuel Ratio for an Effective Combustion of Monticellite and Investigation of Its Microbial and Hemolytic Activity for Biomedical Applications. ACS OMEGA 2023; 8:36919-36932. [PMID: 37841139 PMCID: PMC10568587 DOI: 10.1021/acsomega.3c03984] [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: 06/06/2023] [Accepted: 08/16/2023] [Indexed: 10/17/2023]
Abstract
Bioactive silicates have gained popularity as bone graft substitutes in recent years due to their exceptional ability to bind to host tissues. The current study investigates the effect of changing the metal ion-to-fuel ratio on the properties and biological activity of monticellite prepared via the sol-gel connived combustion technique. Single-phasic monticellite was obtained at 900 °C, without any secondary-phase contaminants for the fuel-lean, stoichiometric, and fuel-rich conditions. SEM and TEM micrographs revealed the porous, spongy morphology of the materials. Because of the reduced crystallite size and higher surface area, the biomineralization of monticellite prepared under fuel-lean conditions resulted in more apatite deposition than those of the other two samples. The results show that the material has a good compressive strength comparable to natural bone, while its brittleness is equivalent to the lower moduli of bone. In terms of antibacterial and antifungal activities, the monticellite bioceramics outperformed the clinical pathogens. It can be used for bone tissue engineering and other biological applications due to its excellent anti-inflammatory and hemolysis inhibitory properties.
Collapse
Affiliation(s)
- Naveensubramaniam Vijayakumar
- Department
of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014, India
| | - Senthil Kumar Venkatraman
- Department
of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014, India
| | - Ravindiran Nandakumar
- Department
of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014, India
| | - Raveena Ann Alex
- Microbial
Biotechnology Laboratory, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Jayanthi Abraham
- Microbial
Biotechnology Laboratory, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Hossein Mohammadi
- Institute
of Energy Infrastructure (IEI), Universiti
Tenaga Nasional, Jalan IKRAMUNITEN, Kajang, Selangor 43000, Malaysia
| | - Mona Ebadi
- Department
of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi, Selangor 43600, Malaysia
| | - Sasikumar Swamiappan
- Department
of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632 014, India
| |
Collapse
|
15
|
Sivakumar PM, Yetisgin AA, Demir E, Sahin SB, Cetinel S. Polysaccharide-bioceramic composites for bone tissue engineering: A review. Int J Biol Macromol 2023; 250:126237. [PMID: 37567538 DOI: 10.1016/j.ijbiomac.2023.126237] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/05/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
Limitations associated with conventional bone substitutes such as autografts, increasing demand for bone grafts, and growing elderly population worldwide necessitate development of unique materials as bone graft substitutes. Bone tissue engineering (BTE) would ensure therapy advancement, efficiency, and cost-effective treatment modalities of bone defects. One way of engineering bone tissue scaffolds by mimicking natural bone tissue composed of organic and inorganic phases is to utilize polysaccharide-bioceramic hybrid composites. Polysaccharides are abundant in nature, and present in human body. Biominerals, like hydroxyapatite are present in natural bone and some of them possess osteoconductive and osteoinductive properties. Ion doped bioceramics could substitute protein-based biosignal molecules to achieve osteogenesis, vasculogenesis, angiogenesis, and stress shielding. This review is a systemic summary on properties, advantages, and limitations of polysaccharide-bioceramic/ion doped bioceramic composites along with their recent advancements in BTE.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Institute of Research and Development, Duy Tan University, Da Nang 550000, Viet Nam; School of Medicine and Pharmacy, Duy Tan University, Da Nang 550000, Viet Nam.
| | - Abuzer Alp Yetisgin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Materials Science and Nano-Engineering Program, Istanbul 34956, Turkey
| | - Ebru Demir
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey
| | - Sevilay Burcu Sahin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey; Sabanci University, Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Istanbul 34956, Turkey.
| |
Collapse
|
16
|
Ivory-Cousins T, Nurzynska A, Klimek K, Baines DK, Truszkiewicz W, Pałka K, Douglas TEL. Whey Protein Isolate/Calcium Silicate Hydrogels for Bone Tissue Engineering Applications-Preliminary In Vitro Evaluation. MATERIALS (BASEL, SWITZERLAND) 2023; 16:6484. [PMID: 37834620 PMCID: PMC10573410 DOI: 10.3390/ma16196484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
Whey protein isolate (WPI) hydrogels are attractive biomaterials for application in bone repair and regeneration. However, their main limitation is low mechanical strength. Therefore, to improve these properties, the incorporation of ceramic phases into hydrogel matrices is currently being performed. In this study, novel whey protein isolate/calcium silicate (WPI/CaSiO3) hydrogel biomaterials were prepared with varying concentrations of a ceramic phase (CaSiO3). The aim of this study was to investigate the effect of the introduction of CaSiO3 to a WPI hydrogel matrix on its physicochemical, mechanical, and biological properties. Our Fourier Transform Infrared Spectroscopy results showed that CaSiO3 was successfully incorporated into the WPI hydrogel matrix to create composite biomaterials. Swelling tests indicated that the addition of 5% (w/v) CaSiO3 caused greater swelling compared to biomaterials without CaSiO3 and ultimate compressive strength and strain at break. Cell culture experiments demonstrated that WPI hydrogel biomaterials enriched with CaSiO3 demonstrated superior cytocompatibility in vitro compared to the control hydrogel biomaterials without CaSiO3. Thus, this study revealed that the addition of CaSiO3 to WPI-based hydrogel biomaterials renders them more promising for bone tissue engineering applications.
Collapse
Affiliation(s)
- Tayla Ivory-Cousins
- School of Engineering, Faculty of Mechanical Engineering, Lancaster University, Nadbystrzycka 36 Street, Gillow Avenue, Lancaster LA1 4YW, UK; (T.I.-C.); (D.K.B.)
| | - Aleksandra Nurzynska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (A.N.); (K.K.); (W.T.)
| | - Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (A.N.); (K.K.); (W.T.)
| | - Daniel K. Baines
- School of Engineering, Faculty of Mechanical Engineering, Lancaster University, Nadbystrzycka 36 Street, Gillow Avenue, Lancaster LA1 4YW, UK; (T.I.-C.); (D.K.B.)
| | - Wieslaw Truszkiewicz
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (A.N.); (K.K.); (W.T.)
| | - Krzysztof Pałka
- Faculty of Mechanical Engineering, Lublin University of Technology, Nadbystrzycka 36 Street, 20-618 Lublin, Poland;
| | - Timothy E. L. Douglas
- School of Engineering, Faculty of Mechanical Engineering, Lancaster University, Nadbystrzycka 36 Street, Gillow Avenue, Lancaster LA1 4YW, UK; (T.I.-C.); (D.K.B.)
| |
Collapse
|
17
|
Kaimonov MR, Safronova TV. Materials in the Na 2O-CaO-SiO 2-P 2O 5 System for Medical Applications. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5981. [PMID: 37687671 PMCID: PMC10488989 DOI: 10.3390/ma16175981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/29/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023]
Abstract
Calcium phosphate materials and materials based on silicon dioxide have been actively studied for more than 50 years due to their high biocompatibility and bioactivity. Hydroxyapatite and tricalcium phosphate are the most known among calcium phosphate materials, and Bioglass 45S5 is the most known material in the Na2O-CaO-SiO2-P2O5 system. Each of these materials has its application limits; however, some of them can be eliminated by obtaining composites based on calcium phosphate and bioglass. In this article, we provide an overview of the role of silicon and its compounds, including Bioglass 45S5, consider calcium phosphate materials, talk about the limits of each material, demonstrate the potential of the composites based on them, and show the other ways of obtaining composite ceramics in the Na2O-CaO-SiO2-P2O5 system.
Collapse
Affiliation(s)
- Maksim R. Kaimonov
- Department of Materials Science, Lomonosov Moscow State University, Leninskie Gory 1, Building 73, 119991 Moscow, Russia
| | - Tatiana V. Safronova
- Department of Materials Science, Lomonosov Moscow State University, Leninskie Gory 1, Building 73, 119991 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, Building 3, 119991 Moscow, Russia
| |
Collapse
|
18
|
Burdusel AC, Neacsu IA, Birca AC, Chircov C, Grumezescu AM, Holban AM, Curutiu C, Ditu LM, Stan M, Andronescu E. Microwave-Assisted Hydrothermal Treatment of Multifunctional Substituted Hydroxyapatite with Prospective Applications in Bone Regeneration. J Funct Biomater 2023; 14:378. [PMID: 37504872 PMCID: PMC10381662 DOI: 10.3390/jfb14070378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Orthopedic bone graft infections are major complications in today's medicine, and the demand for antibacterial treatments is expanding because of the spread of antibiotic resistance. Various compositions of hydroxyapatite (HAp) in which Calcium (Ca2+) ions are substituted with Cerium (Ce3+) and Magnesium (Mg2+) are herein proposed as biomaterials for hard tissue implants. This approach gained popularity in recent years and, in the pursuit of mimicking the natural bone mineral's composition, over 70 elements of the Periodic Table were already reported as substituents into HAp structure. The current study aimed to create materials based on HAp, Hap-Ce, and Hap-Mg using hydrothermal maturation in the microwave field. This route has been considered a novel, promising, and effective way to obtain monodisperse, fine nanoparticles while easily controlling the synthesis parameters. The synthesized HAp powders were characterized morphologically and structurally by XRD diffraction, Dynamic light scattering, zeta potential, FTIR spectrometry, and SEM analysis. Proliferation and morphological analysis on osteoblast cell cultures were used to demonstrate the cytocompatibility of the produced biomaterials. The antimicrobial effect was highlighted in the synthesized samples, especially for hydroxyapatite substituted with cerium. Therefore, the samples of HAp substituted with cerium or magnesium are proposed as biomaterials with enhanced osseointegration, also having the capacity to reduce device-associated infections.
Collapse
Affiliation(s)
- Alexandra-Cristina Burdusel
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
| | - Ionela Andreea Neacsu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
| | - Alexandra Catalina Birca
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
| | - Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
| | - Alexandru-Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Alina Maria Holban
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 077206 Bucharest, Romania
| | - Carmen Curutiu
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 077206 Bucharest, Romania
| | - Lia Mara Ditu
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 077206 Bucharest, Romania
| | - Miruna Stan
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Street, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Splaiul Independentei 54, 050044 Bucharest, Romania
| |
Collapse
|
19
|
Gharbi A, Oudadesse H, El Feki H, Cheikhrouhou-Koubaa W, Chatzistavrou X, V Rau J, Heinämäki J, Antoniac I, Ashammakhi N, Derbel N. High Boron Content Enhances Bioactive Glass Biodegradation. J Funct Biomater 2023; 14:364. [PMID: 37504859 PMCID: PMC10381889 DOI: 10.3390/jfb14070364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/29/2023] Open
Abstract
Derived Hench bioactive glass (BaG) containing boron (B) is explored in this work as it plays an important role in bone development and regeneration. B was also found to enhance BaG dissociation. However, it is only possible to incorporate a limited amount of B. To increase the amount of B in BaG, bioactive borosilicate glasses (BaG-Bx) were fabricated based on the use of the solution-gelation process (sol-gel). In this work, a high B content (20 wt.%) in BaG, respecting the conditions of bioactivity and biodegradability required by Hench, was achieved for the first time. The capability of BaG-Bx to form an apatite phase was assessed in vitro by immersion in simulated body fluid (SBF). Then, the chemical structure and the morphological changes in the fabricated BaG-Bx (x = 0, 5, 10 and 20) were studied. The formation of hydroxyapatite (HAp) layer was observed with X-ray diffraction (XRD) and infrared (IR) spectroscopy. The presence of HAp layer was confirmed using scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Enhanced bioactivity and chemical stability of BaG-Bx were evaluated with an ion exchange study based on Inductively Coupled Plasma-Optical Emission Spectrometry (ICP-OES) and energy dispersive spectroscopy (EDS). Results indicate that by increasing the concentration of B in BaG-Bx, the crystallization rate and the quality of the newly formed HAp layer on BaG-Bx surfaces can be improved. The presence of B also leads to enhanced degradation of BaGs in SBF. Accordingly, BAG-Bx can be used for bone regeneration, especially in children, because of its faster degradation as compared to B-free glass.
Collapse
Affiliation(s)
- Amina Gharbi
- CEM Lab, National Engineering School of Sfax, Sfax University, Sfax 3018, Tunisia
- LT2S Lab, Digital Research Centre of Sfax, Technopole of Sfax, P.O. Box 275, Sfax 3000, Tunisia
| | | | - Hafedh El Feki
- Faculty of Sciences of Sfax, Sfax University, Sfax 3018, Tunisia
| | | | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Julietta V Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy
- Department of Analytical, Physical and Colloid Chemistry, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Jyrki Heinämäki
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Iulian Antoniac
- Faculty of Material Science and Engineering, University Politehnica of Bucharest, SIM 313, 060042 Bucharest, Romania
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering, Department of Biomedical Engineering, College of Engineering and College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Nabil Derbel
- CEM Lab, National Engineering School of Sfax, Sfax University, Sfax 3018, Tunisia
| |
Collapse
|
20
|
Alsaikhan F, Mahmoud MZ, Suliman M. Synthesis and characterization of novel denosumab/magnesium-based metal organic frameworks nanocomposite prepared by ultrasonic route as drug delivery system for the treatment of osteoporosis. Front Bioeng Biotechnol 2023; 11:1153969. [PMID: 37324440 PMCID: PMC10266346 DOI: 10.3389/fbioe.2023.1153969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: The metal-organic frameworks (MOF) have shown fascinating possibilities in biomedical applications, and designing a drug delivery system (DDS) based on the MOF is important. This work aimed at developing a suitable DDS based on Denosumab-loaded Metal Organic Framework/Magnesium (DSB@MOF (Mg)) for attenuating osteoarthritis. Materials and Methods: The MOF (Mg) (Mg3(BPT)2(H2O)4) was synthesized using a sonochemical protocol. The efficiency of MOF (Mg) as a DDS was evaluated by loading and releasing DSB as a drug. In addition, the performance of MOF (Mg) was evaluated by releasing Mg ions for bone formation. The MOF (Mg) and DSB@MOF (Mg) cytotoxicity towards the MG63 cells were explored by MTT assay. Results: MOF (Mg) characterized by using XRD, SEM, EDX, TGA, and BET. Drug loading, and releasing experiments proved that DSB was loaded on the MOF (Mg) and approximately 72% DSB was released from it after 8 h. The characterization techniques showed that MOF (Mg) was successfully synthesized with good crystal structure and thermal stability. The result of BET showed that MOF (Mg) had high surface areas and pore volume. This is the reason why its 25.73% DSB was loaded in the subsequent drug-loading experiment. Drug release and ion release experiments indicated DSB@MOF (Mg) had a good controlled release of DSB and Mg ions in solution. Cytotoxicity assay confirmed that the optimum dose of it had excellent biocompatibility and could stimulate the proliferation of MG63 cells as time went on. Conclusion: Due to the high loading amount of DSB and releasing time, DSB@MOF (Mg) can be promising as a suitable candidate for relieving bone pain caused by osteoporosis, with ossification-reinforcing functions.
Collapse
Affiliation(s)
- Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Mustafa Z. Mahmoud
- Department of Radiology and Medical Imaging, College of Applied Medical Sciences in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
21
|
Ganesh SS, Anushikaa R, Swetha Victoria VS, Lavanya K, Shanmugavadivu A, Selvamurugan N. Recent Advancements in Electrospun Chitin and Chitosan Nanofibers for Bone Tissue Engineering Applications. J Funct Biomater 2023; 14:jfb14050288. [PMID: 37233398 DOI: 10.3390/jfb14050288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/07/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023] Open
Abstract
Treatment of large segmental bone loss caused by fractures, osteomyelitis, and non-union results in expenses of around USD 300,000 per case. Moreover, the worst-case scenario results in amputation in 10% to 14.5% of cases. Biomaterials, cells, and regulatory elements are employed in bone tissue engineering (BTE) to create biosynthetic bone grafts with effective functionalization that can aid in the restoration of such fractured bones, preventing amputation and alleviating expenses. Chitin (CT) and chitosan (CS) are two of the most prevalent natural biopolymers utilized in the fields of biomaterials and BTE. To offer the structural and biochemical cues for augmenting bone formation, CT and CS can be employed alone or in combination with other biomaterials in the form of nanofibers (NFs). When compared with several fabrication methods available to produce scaffolds, electrospinning is regarded as superior since it enables the development of nanostructured scaffolds utilizing biopolymers. Electrospun nanofibers (ENFs) offer unique characteristics, including morphological resemblance to the extracellular matrix, high surface-area-to-volume ratio, permeability, porosity, and stability. This review elaborates on the recent strategies employed utilizing CT and CS ENFs and their biocomposites in BTE. We also summarize their implementation in supporting and delivering an osteogenic response to treat critical bone defects and their perspectives on rejuvenation. The CT- and CS-based ENF composite biomaterials show promise as potential constructions for bone tissue creation.
Collapse
Affiliation(s)
- S Shree Ganesh
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Ramprasad Anushikaa
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Venkadesan Sri Swetha Victoria
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Krishnaraj Lavanya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| |
Collapse
|
22
|
Vijayakumar N, Venkatraman SK, Imthiaz S, Drweesh EA, Elnagar MM, Koppala S, Swamiappan S. Synthesis and characterization of calcium and magnesium based oxides and titanates for photocatalytic degradation of rhodamine B: a comparative study. Sci Rep 2023; 13:3615. [PMID: 36869055 PMCID: PMC9984380 DOI: 10.1038/s41598-023-30013-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
The current investigation deals with the simple and ecological synthesis of CaO, MgO, CaTiO3, and MgTiO3 for the photocatalytic dilapidation of rhodamine B dye. CaO was procured from chicken eggshell waste by calcination process, while MgO was produced by solution combustion method using urea as a fuel source. Furthermore, CaTiO3 and MgTiO3 were synthesized through an easy and simple solid-state method by mixing thoroughly the synthesized CaO or MgO with TiO2 before calcination at 900 °C. XRD and EDX investigations confirmed the phase formation of the materials. Moreover, FTIR spectra revealed the existence of Ca-Ti-O, Mg-Ti-O, and Ti-O which resembles the chemical composition of the proposed materials. SEM micrographs revealed that the surface of CaTiO3 is rougher with relatively dispersed particles compared to MgTiO3, reflecting a higher surface area of CaTiO3. Diffuse reflectance spectroscopy investigations indicated that the synthesized materials can act as photocatalysts under UV illumination. Accordingly, CaO and CaTiO3 effectively degraded rhodamine B dye within 120 min with a photodegradation activity of 63% and 72%, respectively. In contrast, the photocatalytic degradation activity of MgO and MgTiO3 was much lower, since only 21.39 and 29.44% of the dye were degraded, respectively after 120 min of irradiation. Furtheremore, the photocatalytic activity of the mixture from both Ca and Mg titanates was 64.63%. These findings might be valuable for designing potential and affordable photocatalysts for wastewater purification.
Collapse
Affiliation(s)
- Naveensubramaniam Vijayakumar
- grid.412813.d0000 0001 0687 4946Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632014 India
| | - Senthil Kumar Venkatraman
- grid.412813.d0000 0001 0687 4946Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632014 India
| | - Syed Imthiaz
- grid.412813.d0000 0001 0687 4946Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632014 India
| | - Elsayed A. Drweesh
- grid.419725.c0000 0001 2151 8157Department of Inorganic Chemistry, National Research Centre, 33 El Bohouth St. (Former EltahrirSt.), Dokki, Giza, 12622 Egypt
| | - Mohamed M. Elnagar
- grid.419725.c0000 0001 2151 8157Department of Inorganic Chemistry, National Research Centre, 33 El Bohouth St. (Former EltahrirSt.), Dokki, Giza, 12622 Egypt
| | - Sivasankar Koppala
- grid.218292.20000 0000 8571 108XFaculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650093 China
| | - Sasikumar Swamiappan
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
23
|
Pouroutzidou GK, Papadopoulou L, Lazaridou M, Tsachouridis K, Papoulia C, Patsiaoura D, Tsamesidis I, Chrissafis K, Vourlias G, Paraskevopoulos KM, Anastasiou AD, Bikiaris DN, Kontonasaki E. Composite PLGA–Nanobioceramic Coating on Moxifloxacin-Loaded Akermanite 3D Porous Scaffolds for Bone Tissue Regeneration. Pharmaceutics 2023; 15:pharmaceutics15030819. [PMID: 36986685 PMCID: PMC10053907 DOI: 10.3390/pharmaceutics15030819] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/18/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Silica-based ceramics doped with calcium and magnesium have been proposed as suitable materials for scaffold fabrication. Akermanite (Ca2MgSi2O7) has attracted interest for bone regeneration due to its controllable biodegradation rate, improved mechanical properties, and high apatite-forming ability. Despite the profound advantages, ceramic scaffolds provide weak fracture resistance. The use of synthetic biopolymers such as poly(lactic-co-glycolic acid) (PLGA) as coating materials improves the mechanical performance of ceramic scaffolds and tailors their degradation rate. Moxifloxacin (MOX) is an antibiotic with antimicrobial activity against numerous aerobic and anaerobic bacteria. In this study, silica-based nanoparticles (NPs) enriched with calcium and magnesium, as well as copper and strontium ions that induce angiogenesis and osteogenesis, respectively, were incorporated into the PLGA coating. The aim was to produce composite akermanite/PLGA/NPs/MOX-loaded scaffolds through the foam replica technique combined with the sol–gel method to improve the overall effectiveness towards bone regeneration. The structural and physicochemical characterizations were evaluated. Their mechanical properties, apatite forming ability, degradation, pharmacokinetics, and hemocompatibility were also investigated. The addition of NPs improved the compressive strength, hemocompatibility, and in vitro degradation of the composite scaffolds, resulting in them keeping a 3D porous structure and a more prolonged release profile of MOX that makes them promising for bone regeneration applications.
Collapse
Affiliation(s)
- Georgia K. Pouroutzidou
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence: (G.K.P.); (E.K.)
| | - Lambrini Papadopoulou
- School of Geology, Faculty of Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Lazaridou
- Faculty of Sciences, School of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Tsachouridis
- Department of Chemical Engineering and Analytical Science, University of Manchester, Manchester M1 3AL, UK
| | - Chrysanthi Papoulia
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Dimitra Patsiaoura
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Tsamesidis
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Chrissafis
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - George Vourlias
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos M. Paraskevopoulos
- Advanced Materials and Devices Laboratory, Faculty of Sciences, School of Physics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Antonios D. Anastasiou
- Department of Chemical Engineering and Analytical Science, University of Manchester, Manchester M1 3AL, UK
| | - Dimitrios N. Bikiaris
- Faculty of Sciences, School of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Eleana Kontonasaki
- Department of Prosthodontics, Faculty of Health Sciences, School of Dentistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence: (G.K.P.); (E.K.)
| |
Collapse
|
24
|
Nanocomposite Hydrogels as Functional Extracellular Matrices. Gels 2023; 9:gels9020153. [PMID: 36826323 PMCID: PMC9957407 DOI: 10.3390/gels9020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Over recent years, nano-engineered materials have become an important component of artificial extracellular matrices. On one hand, these materials enable static enhancement of the bulk properties of cell scaffolds, for instance, they can alter mechanical properties or electrical conductivity, in order to better mimic the in vivo cell environment. Yet, many nanomaterials also exhibit dynamic, remotely tunable optical, electrical, magnetic, or acoustic properties, and therefore, can be used to non-invasively deliver localized, dynamic stimuli to cells cultured in artificial ECMs in three dimensions. Vice versa, the same, functional nanomaterials, can also report changing environmental conditions-whether or not, as a result of a dynamically applied stimulus-and as such provide means for wireless, long-term monitoring of the cell status inside the culture. In this review article, we present an overview of the technological advances regarding the incorporation of functional nanomaterials in artificial extracellular matrices, highlighting both passive and dynamically tunable nano-engineered components.
Collapse
|
25
|
Pang S, Wu D, Yang H, Kamutzki F, Kurreck J, Gurlo A, Hanaor DAH. Enhanced mechanical performance and bioactivity in strontium/copper co-substituted diopside scaffolds. BIOMATERIALS ADVANCES 2023; 145:213230. [PMID: 36527963 DOI: 10.1016/j.bioadv.2022.213230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/01/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022]
Affiliation(s)
- Shumin Pang
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Dongwei Wu
- Technische Universität Berlin, Chair of Applied Biochemistry, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Haotian Yang
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Franz Kamutzki
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Jens Kurreck
- Technische Universität Berlin, Chair of Applied Biochemistry, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Aleksander Gurlo
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Dorian A H Hanaor
- Technische Universität Berlin, Chair of Advanced Ceramic Materials, Straße des 17. Juni 135, 10623 Berlin, Germany.
| |
Collapse
|
26
|
Blinova AA, Karamirzoev AA, Guseynova AR, Maglakelidze DG, Ilyaeva TA, Gusov BA, Meliksetyants AP, Pirumian MM, Taravanov MA, Pirogov MA, Vakalov DS, Bernyukevich TV, Gvozdenko AA, Nagdalian AA, Blinov AV. Synthesis and Characterization of Calcium Silicate Nanoparticles Stabilized with Amino Acids. MICROMACHINES 2023; 14:245. [PMID: 36837945 PMCID: PMC9967975 DOI: 10.3390/mi14020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
This work presents the development of a method for the synthesis of calcium silicate nanoparticles stabilized with essential amino acids. CaSiO3 nanoparticles were obtained through chemical precipitation. In the first stage, the optimal calcium-containing precursor was determined. The samples were examined using scanning electron microscopy. It was found that Ca(CH3COO)2 was the optimal calcium-containing precursor. Then, the phase composition of calcium silicate was studied using X-ray phase analysis. The results showed the presence of high-intensity bands in the diffractogram, which characterized the phase of the nanosized CaSiO3-wollastonite. In the next stage, the influence of the type of amino acid on the microstructure of calcium silicate was studied. The amnio acids studied were valine, L-leucine, L-isoleucine, L-methionine, L-threonine, L-lysine, L-phenylalanine, and L-tryptophan. The analysis of the SEM micrographs showed that the addition of amino acids did not significantly affect the morphology of the CaSiO3 samples. The surface of the CaSiO3 samples, both without a stabilizer and with amino acids, was represented by irregularly shaped aggregates consisting of nanoparticles with a diameter of 50-400 nm. Further, in order to determine the optimal amino acid to use to stabilize nanoparticles, computerized quantum chemical modeling was carried out. Analysis of the data obtained showed that the most energetically favorable interaction was the CaSiO3-L-methionine configuration, where the interaction occurs through the amino group of the amino acid; the energy value of which was -2058.497 kcal/mol. To confirm the simulation results, the samples were examined using IR spectroscopy. An analysis of the results showed that the interaction of calcium silicate with L-methionine occurs via the formation of a bond through the NH3+ group of the amino acid.
Collapse
Affiliation(s)
- Anastasiya A. Blinova
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | | | - Asiyat R. Guseynova
- Faculty of Dentistry, Derzhavin Tambov State University, 392008 Tambov, Russia
| | - David G. Maglakelidze
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Tatiana A. Ilyaeva
- Faculty of Medicine, Stavropol State Medical University, 355017 Stavropol, Russia
| | - Batradz A. Gusov
- Faculty of Dentistry, North Ossetian State Medical University, 362025 Vladikavkaz, Russia
| | | | - Mari M. Pirumian
- Medical and Preventive Faculty, Rostov State Medical University, 344022 Rostov-on-Don, Russia
| | - Maxim A. Taravanov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Maxim A. Pirogov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Dmitriy S. Vakalov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | | | - Alexey A. Gvozdenko
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Andrey A. Nagdalian
- Laboratory of Food and Industrial Biotechnology, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Andrey V. Blinov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| |
Collapse
|
27
|
Cui J, Yu X, Shen Y, Sun B, Guo W, Liu M, Chen Y, Wang L, Zhou X, Shafiq M, Mo X. Electrospinning Inorganic Nanomaterials to Fabricate Bionanocomposites for Soft and Hard Tissue Repair. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:204. [PMID: 36616113 PMCID: PMC9823959 DOI: 10.3390/nano13010204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Tissue engineering (TE) has attracted the widespread attention of the research community as a method of producing patient-specific tissue constructs for the repair and replacement of injured tissues. To date, different types of scaffold materials have been developed for various tissues and organs. The choice of scaffold material should take into consideration whether the mechanical properties, biodegradability, biocompatibility, and bioresorbability meet the physiological properties of the tissues. Owing to their broad range of physico-chemical properties, inorganic materials can induce a series of biological responses as scaffold fillers, which render them a good alternative to scaffold materials for tissue engineering (TE). While it is of worth to further explore mechanistic insight into the use of inorganic nanomaterials for tissue repair, in this review, we mainly focused on the utilization forms and strategies for fabricating electrospun membranes containing inorganic components based on electrospinning technology. A particular emphasis has been placed on the biological advantages of incorporating inorganic materials along with organic materials as scaffold constituents for tissue repair. As well as widely exploited natural and synthetic polymers, inorganic nanomaterials offer an enticing platform to further modulate the properties of composite scaffolds, which may help further broaden the application prospect of scaffolds for TE.
Collapse
Affiliation(s)
- Jie Cui
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiao Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yihong Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Binbin Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Wanxin Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mingyue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yujie Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Li Wang
- College of Science, Donghua University, Shanghai 201620, China
| | - Xingping Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Muhammad Shafiq
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka 819-0395, Japan
- Department of Biotechnology, Faculty of Science and Technology (FOST), University of Central Punjab (UCP), Lahore 54000, Pakistan
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
28
|
Existing and Novel Biomaterials for Bone Tissue Engineering. Int J Mol Sci 2022; 24:ijms24010529. [PMID: 36613972 PMCID: PMC9820083 DOI: 10.3390/ijms24010529] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
The treatment of bone defects remains one of the major challenges in modern clinical practice. Nowadays, with the increased incidence of bone disease in an aging population, the demand for materials to repair bone defects continues to grow. Recent advances in the development of biomaterials offer new possibilities for exploring modern bone tissue engineering strategies. Both natural and synthetic biomaterials have been used for tissue repair. A variety of porous structures that promote cell adhesion, differentiation, and proliferation enable better implant integration with increasingly better physical properties. The selection of a suitable biomaterial on which the patient's new tissue will grow is one of the key issues when designing a modern tissue scaffold and planning the entire treatment process. The purpose of this article is to present a comprehensive literature review of existing and novel biomaterials used in the surgical treatment of bone tissue defects. The materials described are divided into three groups-organic, inorganic, and synthetic polymers-taking into account current trends. This review highlights different types of existing and novel natural and synthetic materials used in bone tissue engineering and their advantages and disadvantages for bone defects regeneration.
Collapse
|
29
|
Furko M, Horváth ZE, Czömpöly O, Balázsi K, Balázsi C. Biominerals Added Bioresorbable Calcium Phosphate Loaded Biopolymer Composites. Int J Mol Sci 2022; 23:ijms232415737. [PMID: 36555378 PMCID: PMC9779388 DOI: 10.3390/ijms232415737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Nanocrystalline calcium phosphate (CP) bioceramic coatings and their combination with biopolymers are innovative types of resorbable coatings for load-bearing implants that can promote the integration of metallic implants into human bodies. The nanocrystalline, amorphous CP particles are an advantageous form of the various calcium phosphate phases since they have a faster dissolution rate than that of crystalline hydroxyapatite. Owing to the biomineral additions (Mg, Zn, Sr) in optimized concentrations, the base CP particles became more similar to the mineral phase in human bones (dCP). The effect of biomineral addition into the CaP phases was thoroughly studied. The results showed that the shape, morphology, and amorphous characteristic slightly changed in the case of biomineral addition in low concentrations. The optimized dCP particles were then incorporated into a chosen polycaprolactone (PCL) biopolymer matrix. Very thin, non-continuous, rough layers were formed on the surface of implant substrates via the spin coating method. The SEM elemental mapping proved the perfect incorporation and distribution of dCP particles into the polymer matrix. The bioresorption rate of thin films was followed by corrosion measurements over a long period of time. The corrosion results indicated a faster dissolution rate for the dCP-PCL composite compared to the dCP and CP powder layers.
Collapse
|
30
|
Liu J, Tan Y, Shen E, Liu B, Tian Y, Liang L, Yan X, Wu H. Highly water-stable bimetallic organic framework MgCu-MOF74 for inhibiting bacterial infection and promoting bone regeneration. Biomed Mater 2022; 17. [PMID: 36368050 DOI: 10.1088/1748-605x/aca24c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/11/2022] [Indexed: 11/13/2022]
Abstract
As a typical metal-organic framework (MOF), Mg-MOF74 can release biocompatible Mg2+when the framework is degraded, and it has the potential to be used as filler in the field of bone tissue engineering. However, Mg-MOF74 has poor stability in aqueous environment and limited antibacterial ability, which limit its further development and applications. In this work, MgCu-MOF74 particles with different Cu content were synthesized through a facile one-step hydrothermal method. The physicochemical properties and water stability of the synthesized powders were characterized. The osteogenic potential of the MgCu-MOF74 particles on human osteogenic sarcoma cells (SaOS-2) was evaluated. The hybrid MgCu-MOF74 exhibited favorable water stability. These results indicated that MgCu-MOF74 enhanced cellular viability, alkaline phosphatase levels, collagen (COL) synthesis and osteogenesis-related gene expression. Moreover, the samples doped with Cu2+were more sensitive to the acidic microenvironment produced by bacteria, and exhibited stronger antibacterial ability than Mg-MOF74. In conclusion, MgCu-MOF-74 with good water stability, osteogenic ability and antibacterial ability, which could be attributed to the doping of Cu2+. Hence, MgCu-MOF74 shows great potential as a novel medical bio-functional fillers for the treatment of bone defects.
Collapse
Affiliation(s)
- Jiamin Liu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, People's Republic of China
| | - Yanni Tan
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, People's Republic of China
| | - Erdong Shen
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, People's Republic of China.,Department of Oncology, Yueyang Central Hospital, Yueyang 414000, People's Republic of China
| | - Bo Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Yingtao Tian
- Department of Engineering, Lancaster University, Bailrigg, Lancaster LA1 4YW, United Kingdom
| | - Luxin Liang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Xinxin Yan
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan 430060, People's Republic of China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, People's Republic of China
| |
Collapse
|
31
|
Bharathi R, Ganesh SS, Harini G, Vatsala K, Anushikaa R, Aravind S, Abinaya S, Selvamurugan N. Chitosan-based scaffolds as drug delivery systems in bone tissue engineering. Int J Biol Macromol 2022; 222:132-153. [PMID: 36108752 DOI: 10.1016/j.ijbiomac.2022.09.058] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
Abstract
The bone tissue engineering approach for treating large bone defects becomes necessary when the tissue damage surpasses the threshold of the inherent regenerative ability of the human body. A myriad of natural biodegradable polymers and scaffold fabrication techniques have emerged in the last decade. Chitosan (CS) is especially attractive as a bone scaffold material to support cell attachment and proliferation and mineralization of the bone matrix. The primary amino groups in CS are responsible for properties such as controlled drug release, mucoadhesion, in situ gelation, and transfection. CS-based smart drug delivery scaffolds that respond to environmental stimuli have been reported to have a localized sustained delivery of drugs in the large bone defect area. This review outlines the recent advances in the fabrication of CS-based scaffolds as a pharmaceutical carrier to deliver drugs such as antibiotics, growth factors, nucleic acids, and phenolic compounds for bone tissue regeneration.
Collapse
Affiliation(s)
- R Bharathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - S Shree Ganesh
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - G Harini
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Kumari Vatsala
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - R Anushikaa
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - S Aravind
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - S Abinaya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
32
|
Guo E, Wu J, Lu H, Wang L, Chen Q. Tissue-engineered bones with adipose-derived stem cells - composite polymer for repair of bone defects. Regen Med 2022; 17:643-657. [PMID: 35703025 DOI: 10.2217/rme-2022-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Development of alternative bone tissue graft materials based on tissue engineering technology has gradually become a research focus. Engineered bone composed of biodegradable, biosafe and bioactive materials is attractive, but also challenging. Materials & methods: An adipose-derived stem cell/poly(L-glutamic acid)/chitosan composite scaffold was further developed for construction of biodegradable and bone-promoting tissue-engineered bone. A series of composite scaffold materials with different physical properties such as structure, pore size, porosity and pore diameter was developed. Results: The composite scaffold showed good biodegradability and water absorption, and exhibited an excellent ability to promote bone differentiation. Conclusion: This type of biodegradable scaffold is expected to be applied to the field of bone repair or bone tissue engineering.
Collapse
Affiliation(s)
- Enqi Guo
- Department of Hand & Reconstructive Surgery, Plastic & Reconstructive Surgery Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jianlong Wu
- Department of Hand & Reconstructive Surgery, Plastic & Reconstructive Surgery Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Hongrui Lu
- Department of Hand & Reconstructive Surgery, Plastic & Reconstructive Surgery Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Liang Wang
- Department of Hand & Reconstructive Surgery, Plastic & Reconstructive Surgery Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Qiang Chen
- Department of Hand & Reconstructive Surgery, Plastic & Reconstructive Surgery Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
33
|
PCL/Si-Doped Multi-Phase Calcium Phosphate Scaffolds Derived from Cuttlefish Bone. MATERIALS 2022; 15:ma15093348. [PMID: 35591682 PMCID: PMC9102552 DOI: 10.3390/ma15093348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023]
Abstract
Increasing attention is focused on developing biomaterials as temporary scaffolds that provide a specific environment and microstructure for bone tissue regeneration. The aim of the present work was to synthesize silicon-doped biomimetic multi-phase composite scaffolds based on bioactive inorganic phases and biocompatible polymers (poly(ε-caprolactone), PCL) using simple and inexpensive methods. Porous multi-phase composite scaffolds from cuttlefish bone were synthesized using a hydrothermal method and were further impregnated with (3-aminopropyl)triethoxysilane 1–4 times, heat-treated (1000 °C) and coated with PCL. The effect of silicon doping and the PCL coating on the microstructure and mechanical and biological properties of the scaffolds has been investigated. Multi-phase scaffolds based on calcium phosphate (hydroxyapatite, α-tricalcium phosphate, β-tricalcium phosphate) and calcium silicate (wollastonite, larnite, dicalcium silicate) phases were obtained. Elemental mapping revealed homogeneously dispersed silicon throughout the scaffolds, whereas silicon doping increased bovine serum albumin protein adsorption. The highly porous structure of cuttlefish bone was preserved with a composite scaffold porosity of ~78%. A compressive strength of ~1.4 MPa makes the obtained composite scaffolds appropriate for non-load-bearing applications. Cytocompatibility assessment by an MTT assay of human mesenchymal stem cells revealed the non-cytotoxicity of the obtained scaffolds.
Collapse
|
34
|
Li Y, Wang X. Chrysin Attenuates High Glucose-Induced BMSC Dysfunction via the Activation of the PI3K/AKT/Nrf2 Signaling Pathway. Drug Des Devel Ther 2022; 16:165-182. [PMID: 35058687 PMCID: PMC8763623 DOI: 10.2147/dddt.s335024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose High glucose environment in diabetes mellitus induces the dysfunction of bone marrow-derived mesenchymal stromal cells (BMSCs) and impairs bone regeneration. Chrysin is a natural polyphenol with outstanding anti-inflammation and anti-oxidation ability. However, whether and how chrysin affects BMSCs in high glucose conditions remain poorly understood. The present study aimed to explore the effects and underlying mechanisms of chrysin on the BMSCs exposed to high glucose environment. Materials and Methods Cell viability was detected by cell counting kit 8 assay and 5-ethynyl-2’-deoxyuridine staining, while cell apoptosis was determined through flow cytometry using Annexin V-FITC/PI kit. The oxidative stress in BMSCs was evaluated by detecting the reactive oxygen species production, malondialdehyde content, and superoxide dismutase activity. Alkaline phosphatase staining, Alizarin Red staining, and quantitative real-time PCR were performed to determine the osteogenic differentiation. Western blot was used to examine the expression of the PI3K/ATK/Nrf2 signaling pathway. Furthermore, chrysin was injected into calvarial defects of type 1 diabetic SD rats to assess its in vivo bone formation capability. Results Chrysin reduced oxidative stress, increased cell viability, and promoted osteogenic differentiation in BMSCs exposed to high glucose. Blocking PI3K/ATK/Nrf2 signaling pathway weakened the beneficial effects of chrysin, indicating that chrysin at least partly worked through the PI3K/ATK/Nrf2 pathway. Conclusion Chrysin can protect BMSCs from high glucose-induced oxidative stress via the activation of the PI3K/AKT/Nrf2 pathway, and promote bone regeneration in type 1 diabetic rats.
Collapse
Affiliation(s)
- Yu Li
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Department of Hand, Plastic and Reconstructive Surgery, Burn Center-Hand and Plastic Surgery, University of Heidelberg, BG Trauma Center Ludwigshafen, Ludwigshafen, Germany
| | - Ximei Wang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
35
|
Salamanca E, Pan YH, Sun YS, Hsueh HW, Dorj O, Yao WL, Lin JCY, Teng NC, Watanabe I, Abe S, Wu YF, Chang WJ. Magnesium Modified β-Tricalcium Phosphate Induces Cell Osteogenic Differentiation In Vitro and Bone Regeneration In Vivo. Int J Mol Sci 2022; 23:ijms23031717. [PMID: 35163639 PMCID: PMC8836187 DOI: 10.3390/ijms23031717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 01/27/2023] Open
Abstract
In vitro, in vivo, and clinical studies have shown how the physicochemical and biological properties of β-tricalcium phosphate (β-TCP) work in bone regeneration. This study aimed to improve the properties of β-TCP by achieving optimum surface and bulk β-TCP chemical/physical properties through the hydrothermal addition of magnesium (Mg) and to later establish the biocompatibility of β-TCP/Mg for bone grafting and tissue engineering treatments. Multiple in vitro and in vivo analyses were used to complete β-TCP/Mg physicochemical and biological characterization. The addition of MgO brought about a modest rise in the number of β-TCP surface particles, indicating improvements in alkaline phosphatase (ALP) activity on day 21 (p < 0.05) and in the WST-1assay on all days (p < 0.05), with a corresponding increase in the upregulation of ALP and bone sialoprotein. SEM analyses stated that the surfaces of the β-TCP particles were not altered after the addition of Mg. Micro-CT and histomorphometric analysis from rabbit calvaria critical defects resulted in β-TCP/Mg managing to reform more new bone than the control defects and β-TCP control at 2, 6, and 8 weeks (* p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, and **** p ≤ 0.0001). The hydrothermal addition of MgO to the β-TCP surfaces ameliorated its biocompatibility without altering its surface roughness resulting from the elemental composition while enhancing cell viability and proliferation, inducing more bone regeneration by osteoconduction in vivo and osteoblastic differentiation in vitro.
Collapse
Affiliation(s)
- Eisner Salamanca
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
| | - Yu-Hwa Pan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Department of General Dentistry, Chang Gung Memorial Hospital, Taipei 10507, Taiwan
- Graduate Institute of Dental & Craniofacial Science, Chang Gung University, Taoyuan 33305, Taiwan
- School of Dentistry, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Ying-Sui Sun
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Hao-Wen Hsueh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
| | - Odontuya Dorj
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Department of Dental Technology and Hygiene, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia
| | - Wan-Ling Yao
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
| | - Jerry Chin-Yi Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental, Medicine, Boston, MA 02115, USA
| | - Nai-Chia Teng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Dental Department, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Ikki Watanabe
- Department of Gerontology, Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, Tokyo 101-0061, Japan;
| | - Yi-Fan Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Correspondence: (Y.-F.W.); (W.-J.C.); Tel.: +886-2-2736-1661 (ext. 5148) (Y.-F.W.); +886-2-2736-1661 (ext. 5150) (W.-J.C.)
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan; (E.S.); (Y.-H.P.); (H.-W.H.); (O.D.); (W.-L.Y.); (J.C.-Y.L.); (N.-C.T.)
- Dental Department, Shuang-ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Correspondence: (Y.-F.W.); (W.-J.C.); Tel.: +886-2-2736-1661 (ext. 5148) (Y.-F.W.); +886-2-2736-1661 (ext. 5150) (W.-J.C.)
| |
Collapse
|
36
|
Deng Y, Wei W, Tang P. Applications of Calcium-Based Nanomaterials in Osteoporosis Treatment. ACS Biomater Sci Eng 2022; 8:424-443. [PMID: 35080365 DOI: 10.1021/acsbiomaterials.1c01306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With rapidly aging populations worldwide, osteoporosis has become a serious global public health problem. Caused by disordered systemic bone remodeling, osteoporosis manifests as progressive loss of bone mass and microarchitectural deterioration of bone tissue, increasing the risk of fractures and eventually leading to osteoporotic fragility fractures. As fracture risk increases, antiosteoporosis treatments transition from nonpharmacological management to pharmacological intervention, and finally to the treatment of fragility fractures. Calcium-based nanomaterials (CBNMs) have unique advantages in osteoporosis treatment because of several characteristics including similarity to natural bone, excellent biocompatibility, easy preparation and functionalization, low pH-responsive disaggregation, and inherent pro-osteogenic properties. By combining additional ingredients, CBNMs can play multiple roles to construct antiosteoporotic biomaterials with different forms. This review covers recent advances in CBNMs for osteoporosis treatment. For ease of understanding, CBNMs for antiosteoporosis treatment can be classified as locally applied CBNMs, such as implant coatings and filling materials for osteoporotic bone regeneration, and systemically administered CBNMs for antiosteoporosis treatment. Locally applied CBNMs for osteoporotic bone regeneration develop faster than the systemically administered CBNMs, an important consideration given the serious outcomes of fragility fractures. Nevertheless, many innovations in construction strategies and preparation methods have been applied to build systemically administered CBNMs. Furthermore, with increasing interest in delaying osteoporosis progression and avoiding fragility fracture occurrence, research into systemic administration of CBNMs for antiosteoporosis treatment will have more development prospects. Deep understanding of the CBNM preparation process and optimizing CBNM properties will allow for increased application of CBNMs in osteoporosis treatments in the future.
Collapse
Affiliation(s)
- Yuan Deng
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences No. 1 Bei-Er-Tiao, Beijing 100190, P. R. China
| | - Peifu Tang
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| |
Collapse
|
37
|
Vijayakumar N, Venkatraman SK, Choudhary R, Indurkar A, Chatterjee A, Abraham J, Ostrovskiy S, Senatov F, Locs J, Swamiappan S. Conversion of Biowaste into Larnite by Sol‐Gel Combustion Route for Biomedical Applications. ChemistrySelect 2022. [DOI: 10.1002/slct.202103783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - Senthil Kumar Venkatraman
- Department of Chemistry Saveetha School of Engineering Saveetha Institute of Medical and Technical Sciences (SIMATS) Chennai, Tamil Nadu 602105 India
| | - Rajan Choudhary
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU Institute of General Chemical Engineering Faculty of Materials Science and Applied Chemistry Riga Technical University Pulka St 3 LV-1007 Riga Latvia
- Baltic Biomaterials Centre of Excellence Headquarters at Riga Technical University Kalku Street 1 LV-1007 Riga Latvia
- Center for Biomedical Engineering National University of Science and Technology “MISiS” Moscow 119049, Leninskiy Prospect 4 Russia
| | - Abhishek Indurkar
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU Institute of General Chemical Engineering Faculty of Materials Science and Applied Chemistry Riga Technical University Pulka St 3 LV-1007 Riga Latvia
- Baltic Biomaterials Centre of Excellence Headquarters at Riga Technical University Kalku Street 1 LV-1007 Riga Latvia
| | - Ankita Chatterjee
- Microbiol Biotechnology Laboratory School of Biosciences and Technology Vellore Institute of Technology Vellore Tamil Nadu 632014 India
| | - Jayanti Abraham
- Microbiol Biotechnology Laboratory School of Biosciences and Technology Vellore Institute of Technology Vellore Tamil Nadu 632014 India
| | - Sergey Ostrovskiy
- Center for Biomedical Engineering National University of Science and Technology “MISiS” Moscow 119049, Leninskiy Prospect 4 Russia
| | - Fedor Senatov
- Center for Biomedical Engineering National University of Science and Technology “MISiS” Moscow 119049, Leninskiy Prospect 4 Russia
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU Institute of General Chemical Engineering Faculty of Materials Science and Applied Chemistry Riga Technical University Pulka St 3 LV-1007 Riga Latvia
- Baltic Biomaterials Centre of Excellence Headquarters at Riga Technical University Kalku Street 1 LV-1007 Riga Latvia
| | - Sasikumar Swamiappan
- Department of Chemistry School of Advanced Sciences Vellore Institute of Technology Vellore Tamil Nadu 632014 India
| |
Collapse
|
38
|
Song Y, Xu L, Jin X, Chen D, Jin X, Xu G. Effect of calcium and magnesium on inflammatory cytokines in accidentally multiple fracture adults: A short-term follow-up. Medicine (Baltimore) 2022; 101:e28538. [PMID: 35029924 PMCID: PMC8735799 DOI: 10.1097/md.0000000000028538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca) and magnesium (Mg), which play an important role in several cellular processes, is essential for normal development of the skeleton and maintenance of tissue homeostasis. Deficiency of these elements might delay bone fracture recovery or accelerates bone loss. We aimed to examine whether supplementation of trace element (TE) promotes fracture healing in accidentally fracturing adults by involvement of inflammatory mechanism.A short-term follow-up in clinic was performed. Totally, 117 subjects diagnosed with multiple fractures by traffic accidents were recruited in this study. Serum Ca and Mg levels were measured by inductively coupled plasma atomic emission spectrophotometry. Short-term changes such as serum C-reactive protein, interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha in normal treatment and TE supplement groups were detected by enzyme-linked immunosorbent assay. Student t test and the Spearman correlation were performed to analyze the data.Significantly negative correlations between Ca (r = 0.7032; P < .001) and Mg (r = 0.2719; P < .05) and injury severity score were observed. Serum Ca and Mg were significantly increased at Day 5, 7, and 9 following TE supplements. After treatment, serum C-reactive protein, IL-1β, IL-6, and tumor necrosis factor alpha were significantly reduced whereas cytokine levels of the TE supplement group were found to be lower than that of the normal treatment group after Day 3.These findings suggest that Ca and Mg levels are associated with the injury severity of multiple fractures, and the supplement could reduce the inflammation, which may be beneficial for the bone recovery and disease process.
Collapse
Affiliation(s)
- Yongxing Song
- Department of Orthopedics, China Coast Guard Hospital of the People's Armed Police Force, Jiangnan Hospital Affiliated to Jiaxing University, Jiaxing, ZJ, China
| | - Long Xu
- Molecular Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University Medical College, Jiaxing, ZJ, China
| | - Xin Jin
- Molecular Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University Medical College, Jiaxing, ZJ, China
| | - Deqing Chen
- Molecular Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University Medical College, Jiaxing, ZJ, China
| | - Xiuhui Jin
- Department of Immunology and Human Biology, University of Toronto, Toronto, ON, Canada
| | - Guangtao Xu
- Molecular Pathology Laboratory, Department of Pathology, Institute of Forensic Science, Jiaxing University Medical College, Jiaxing, ZJ, China
| |
Collapse
|
39
|
Samoylenko O, Korotych O, Manilo M, Samchenko Y, Shlyakhovenko V, Lebovka N. Biomedical Applications of Laponite®-Based Nanomaterials and Formulations. SPRINGER PROCEEDINGS IN PHYSICS 2022:385-452. [DOI: 10.1007/978-3-030-80924-9_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Ge Y, Wang K, Li H, Tian Y, Wu Y, Lin Z, Lin Y, Wang Y, Zhang J, Tang B. An Mg-MOFs based multifunctional medicine for the treatment of osteoporotic pain. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112386. [PMID: 34579905 DOI: 10.1016/j.msec.2021.112386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/30/2021] [Accepted: 08/17/2021] [Indexed: 11/27/2022]
Abstract
Bone pain is the primary problem for patients with osteoporosis. Ketoprofen is clinically used to treat osteoporotic pain, while long-term oral administration of ketoprofen can cause some side effects. In addition, osteoporosis is also accompanied by bone mass loss and inflammation. In this study, we designed a multifunctional drug (Ket@Mg-MOF-74) adopted Mg-MOF-74 to load ketoprofen to treat osteoporotic pain, bone loss and inflammation comprehensively. Mg-MOF-74 was prepared, and the physicochemical characterization proved that it had excellent physical and chemical stability. Ket@Mg-MOF-74 was synthesized by post-synthetic modification method and a high loading rate of ketoprofen was confirmed. Drug release and ion release experiments indicated Ket@Mg-MOF-74 had a good controlled release of ketoprofen and Mg in solution. Cell experiments in vitro proved the compound drug could significantly reduce the expression of pain-related genes of cyclooxygenase 2 (COX2), obviously up-regulated the expression of osteogenic cytokines and remarkably down-regulated the secretion of pro-inflammatory factors. Therefore, Ket@Mg-MOF-74 is believed a promising painkiller for osteoporotic bone pain, with the function of anti-inflammatory and promoting bone formation.
Collapse
Affiliation(s)
- Yongmei Ge
- Harbin Institute of Technology, Harbin, Heilongjiang 150001, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Kui Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Department of Mechanical Engineering, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Huili Li
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ye Tian
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Yutong Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhaowei Lin
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yangyang Lin
- Department of Rehabilitation Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yansong Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jiarong Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
41
|
Karimzadeh Bardeei L, Seyedjafari E, Hossein G, Nabiuni M, Majles Ara MH, Salber J. Regeneration of Bone Defects in a Rabbit Femoral Osteonecrosis Model Using 3D-Printed Poly (Epsilon-Caprolactone)/Nanoparticulate Willemite Composite Scaffolds. Int J Mol Sci 2021; 22:10332. [PMID: 34638673 PMCID: PMC8508893 DOI: 10.3390/ijms221910332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 01/12/2023] Open
Abstract
Steroid-associated osteonecrosis (SAON) is a chronic disease that leads to the destruction and collapse of bone near the joint that is subjected to weight bearing, ultimately resulting in a loss of hip and knee function. Zn2+ ions, as an essential trace element, have functional roles in improving the immunophysiological cellular environment, accelerating bone regeneration, and inhibiting biofilm formation. In this study, we reconstruct SAON lesions with a three-dimensional (3D)-a printed composite made of poly (epsilon-caprolactone) (PCL) and nanoparticulate Willemite (npW). Rabbit bone marrow stem cells were used to evaluate the cytocompatibility and osteogenic differentiation capability of the PCL/npW composite scaffolds. The 2-month bone regeneration was assessed by a Micro-computed tomography (micro-CT) scan and the expression of bone regeneration proteins by Western blot. Compared with the neat PCL group, PCL/npW scaffolds exhibited significantly increased cytocompatibility and osteogenic activity. This finding reveals a new concept for the design of a 3D-printed PCL/npW composite-based bone substitute for the early treatment of osteonecrosis defects.
Collapse
Affiliation(s)
- Latifeh Karimzadeh Bardeei
- Developmental Biology Laboratory, Animal Biology Department, School of Biology, College of Science, University of Tehran, Tehran 1417935840, Iran;
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran 1417935840, Iran
| | - Ghamartaj Hossein
- Developmental Biology Laboratory, Animal Biology Department, School of Biology, College of Science, University of Tehran, Tehran 1417935840, Iran;
| | - Mohammad Nabiuni
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran;
| | - Mohammad Hosein Majles Ara
- Photonics Laboratory, Physics Department, Kharazmi University, Tehran 15719-14911, Iran;
- Applied Science Research Centre, Kharazmi University, Tehran 15719-14911, Iran
| | - Jochen Salber
- Salber Laboratory, Centre for Clinical Research, Department of Experimental Surgery, Ruhr-Universität Bochum, 44780 Bochum, Germany;
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum GmbH, 44892 Bochum, Germany
| |
Collapse
|
42
|
Genasan K, Mehrali M, Veerappan T, Talebian S, Malliga Raman M, Singh S, Swamiappan S, Mehrali M, Kamarul T, Balaji Raghavendran HR. Calcium-Silicate-Incorporated Gellan-Chitosan Induced Osteogenic Differentiation in Mesenchymal Stromal Cells. Polymers (Basel) 2021; 13:3211. [PMID: 34641027 PMCID: PMC8512901 DOI: 10.3390/polym13193211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
Gellan-chitosan (GC) incorporated with CS: 0% (GC-0 CS), 10% (GC-10 CS), 20% (GC-20 CS) or 40% (GC-40 CS) w/w was prepared using freeze-drying method to investigate its physicochemical, biocompatible, and osteoinductive properties in human bone-marrow mesenchymal stromal cells (hBMSCs). The composition of different groups was reflected in physicochemical analyses performed using BET, FTIR, and XRD. The SEM micrographs revealed excellent hBMSCs attachment in GC-40 CS. The Alamar Blue assay indicated an increased proliferation and viability of seeded hBMSCs in all groups on day 21 as compared with day 0. The hBMSCs seeded in GC-40 CS indicated osteogenic differentiation based on an amplified alkaline-phosphatase release on day 7 and 14 as compared with day 0. These cells supported bone mineralization on GC-40 CS based on Alizarin-Red assay on day 21 as compared with day 7 and increased their osteogenic gene expression (RUNX2, ALP, BGLAP, BMP, and Osteonectin) on day 21. The GC-40 CS-seeded hBMSCs initiated their osteogenic differentiation on day 7 as compared with counterparts based on an increased expression of type-1 collagen and BMP2 in immunocytochemistry analysis. In conclusion, the incorporation of 40% (w/w) calcium silicate in gellan-chitosan showed osteoinduction potential in hBMSCs, making it a potential biomaterial to treat critical bone defects.
Collapse
Affiliation(s)
- Krishnamurithy Genasan
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
| | - Mohammad Mehrali
- Faculty of Engineering Technology, Department of Thermal and Fluid Engineering (TFE), University of Twente, 7500 AE Enschede, The Netherlands;
| | - Tarini Veerappan
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
| | - Sepehr Talebian
- Faculty of Engineering, School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
- Nano Institute (Sydney Nano), The University of Sydney, Sydney, NSW 2006, Australia
| | - Murali Malliga Raman
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
| | - Simmrat Singh
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
| | - Sasikumar Swamiappan
- Materials Chemistry Division, School of Advanced Sciences, VIT University, Vellore 632014, Tamil Nadu, India;
| | - Mehdi Mehrali
- Department of Mechanical Engineering, Technical University of Denmark, 2800 Kgs Lyngby, Denmark;
| | - Tunku Kamarul
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
- Advanced Medical and Dental Institute (AMDI), University Sains Malaysia, Bertam, Kepala Batas 13200, Penang, Malaysia
| | - Hanumantha Rao Balaji Raghavendran
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Tissue Engineering Group (TEG), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (K.G.); (T.V.); (M.M.R.); (S.S.)
- Faculty of Clinical Research, Central Research Facility, Sri Ramachandra Institute of Higher Education and Research Porur, Chennai 600116, Tamil Nadu, India
| |
Collapse
|
43
|
Tonelli M, Faralli A, Ridi F, Bonini M. 3D printable magnesium-based cements towards the preparation of bioceramics. J Colloid Interface Sci 2021; 598:24-35. [PMID: 33892441 DOI: 10.1016/j.jcis.2021.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/28/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
HYPOTHESIS Among all the materials used so far to replace and repair damaged bone tissues, magnesium silicate bioceramics are one of the most promising, thanks to their biocompatibility, osteoinductive properties and good mechanical stability. EXPERIMENTS Magnesium silicate cement pastes were prepared by hydration of MgO mixed with different SiO2 batches at different Mg/Si molar ratios. Pastes were either moulded or 3D printed to obtain set cements that were then calcined at 1000 °C to produce biologically relevant ceramic materials. Both cements and ceramics were characterized by means of X-ray diffraction, while two selected formulations were thoroughly characterized by means of injectability tests, Raman confocal microscopy, scanning electron microscopy, atomic force microscopy, gas porosimetry, X-ray microtomography and compressive tests. FINDINGS The results show that bioceramic scaffolds, namely forsterite and clinoenstatite, can be effectively obtained by 3D printing MgO/SiO2 cement pastes, paving the way towards important advances in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Monica Tonelli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.
| | - Andrea Faralli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.
| | - Francesca Ridi
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.
| | - Massimo Bonini
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
44
|
Bioactive Calcium Phosphate-Based Composites for Bone Regeneration. JOURNAL OF COMPOSITES SCIENCE 2021. [DOI: 10.3390/jcs5090227] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Calcium phosphates (CaPs) are widely accepted biomaterials able to promote the regeneration of bone tissue. However, the regeneration of critical-sized bone defects has been considered challenging, and the development of bioceramics exhibiting enhanced bioactivity, bioresorbability and mechanical performance is highly demanded. In this respect, the tuning of their chemical composition, crystal size and morphology have been the matter of intense research in the last decades, including the preparation of composites. The development of effective bioceramic composite scaffolds relies on effective manufacturing techniques able to control the final multi-scale porosity of the devices, relevant to ensure osteointegration and bio-competent mechanical performance. In this context, the present work provides an overview about the reported strategies to develop and optimize bioceramics, while also highlighting future perspectives in the development of bioactive ceramic composites for bone tissue regeneration.
Collapse
|
45
|
Mosaad KE, Shoueir KR, Saied AH, Dewidar MM. New Prospects in Nano Phased Co-substituted Hydroxyapatite Enrolled in Polymeric Nanofiber Mats for Bone Tissue Engineering Applications. Ann Biomed Eng 2021; 49:2006-2029. [PMID: 34378121 DOI: 10.1007/s10439-021-02810-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/03/2021] [Indexed: 01/12/2023]
Abstract
The most common forms of tissue impairment are fracture bones and significant bone disorders caused by multiple traumas or normal aging. Surgical care sometimes necessitates the placement of a temporary or permanent prosthesis, which continues to be a challenge for orthopedic surgeons, including those with large bone defects. Electrospun scaffolds made from natural and synthetic nanofiber-based polymers are studied as natural extracellular matrix (ECM)-like scaffolds for tissue engineering. Besides, nanostructured materials have properties and functions depending on the scale of natural materials such as hydroxyapatite (HAP), ranging from 1 to 100 nm, which activity was proficient upon enrolled in nanofiber mats. The use of nanofibers in combination with nano-HAP has increased the scaffold's ability to replicate the construction of natural bone tissue that is the aim of the present text. In bone engineering, nanofiber substrates facilitate cell adhesion, proliferation, and differentiation, while HAP induces cells to secrete ECM for bone mineralization and development. This review aims to draw the reader's attention to the critical issues with synthetic and natural polymers containing HAP in bone tissue engineering; co-substituted hydroxyapatite has also been mentioned.
Collapse
Affiliation(s)
- Kareem E Mosaad
- Faculty of Engineering, Mechanical Department, Al-Azahar University, Cairo, Egypt
| | - Kamel R Shoueir
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516, Kafrelsheikh, Egypt.
- Institut de Chimie et Procédés Pour l'Énergie, l'Environnement et la Santé (ICPEES), CNRS, UMR 7515, Université de Strasbourg, 25 rue Becquerel, 67087, Strasbourg, France.
| | - Ahmed H Saied
- Department of Mechanical Engineering, Faculty of Engineering, Kafrelsheikh University, El-Gaish Street, Kafrelsheikh, Egypt
| | - Montasser M Dewidar
- Department of Mechanical Engineering, Faculty of Engineering, Kafrelsheikh University, El-Gaish Street, Kafrelsheikh, Egypt
- Higher Institute of Engineering and Technology, Kafrelsheikh, Egypt
| |
Collapse
|
46
|
Bulanov EN, Petrov SS, Xu Z, Knyazev AV, Skoblikov NE. Synthesis and Crystal Structure of Some Ba-Apatites. RUSS J INORG CHEM+ 2021. [DOI: 10.1134/s0036023621040069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|