1
|
García-Perdiguero JC, Gómez-Cerezo N, Gisbert-Garzarán M, Manzano M, Vallet-Regí M. Unraveling the role of calcium in the osteogenic behavior of mesoporous bioactive glass nanoparticles. Acta Biomater 2025:S1742-7061(25)00247-8. [PMID: 40187671 DOI: 10.1016/j.actbio.2025.04.005] [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: 12/05/2024] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
The use of bioactive materials has emerged as a promising strategy to circumvent bone-related diseases. Because of their chemical composition, calcium-containing bioactive glasses, including mesoporous bioactive glass nanoparticles (nMBG), have long demonstrated their bone regeneration features. In this work, SiO₂-CaO nMBG were synthesized varying Si/Ca ratio from 10 % to 40 % to explore the role of Ca in the osteogenic properties of such materials. We have performed an in-depth physicochemical and biological evaluation of samples by TEM, FTIR, adsorption nitrogen and solid state NMR, revealing that increasing calcium weakens the silica network and consequently, the osteogenic properties. In addition, we have evaluated the protein corona in human serum, obtaining varying protein patterns depending on the Si/Ca ratio and the incubation time. The cellular studies have shown that only certain amounts of calcium up-regulate the osteogenic differentiation, although exceeding such concentrations does not provide improved effects. Finally, All Ca-containing samples promoted calcium phosphate mineralization in biological fluids, while those with higher Si/Ca ratios enhanced significantly hMSC and hOB mineralization. Calcium also modulated hMSC gene expression, with samples containing up to 20 % calcium up-regulating OC and RUNX2. Furthermore, nMBG exhibited immunomodulatory properties, inducing a shift toward the M2 reparative phenotype. Overall, this comprehensive study highlights the crucial role of calcium in osteogenic responses, demonstrating that calcium quantity alone does not surpass the importance of structural and compositional quality in nanosized MBG. STATEMENT OF SIGNIFICANCE: Bone-related diseases are becoming a major socioeconomic issue owing to the increased aging of our society. Therefore, bioactive materials based on silicon, calcium and phosphorus have been used for years due to the osteogenic properties of these elements. In the last few years, the preparation of these materials as nanoparticles has increased their range of applications. In this sense, the novelty of our work relies on the in-depth physicochemical and biological evaluation of those mesoporous bioactive glass nanoparticles based on silicon and calcium, which remained unexplored so far. Couple with the establishment of the range of atomic percentage of calcium with respect to silicon that up-regulate the osteogenic differentiation, although exceeding such concentrations does not provide improved effects.
Collapse
Affiliation(s)
- José C García-Perdiguero
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
| | - Natividad Gómez-Cerezo
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
| | - Miguel Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
| | - Miguel Manzano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain; Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| |
Collapse
|
2
|
Ma H, Tong J, Su X, Liu L, Liang J, Sun J, Lu J, Zhang Y, Lei B, Zhao H. 3D Printed Bioactive Mechanical-Adaptive Polyetheretherketone Implants with Non-Invasive Tracking for Immunomodulatory Osseointegration. Adv Healthc Mater 2025; 14:e2404435. [PMID: 39996270 DOI: 10.1002/adhm.202404435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/30/2024] [Indexed: 02/26/2025]
Abstract
Polyether-ether-ketone (PEEK) has become a much-attracted biomedical implant material in orthopedic surgery, serving as a more biocompatible alternative to conventional metals. However, the inherent bioinert and mismatched mechanical surface of PEEK have limited their optimized bone fixation and repair. In this work, a PEEK implant is printed and a bioactive mechanical-adaptive surface via in situ chemical linking of photoluminescent elastomeric poly(citrate-silicon) (PCS) polymer (PEEK-PCS) is subsequently constructed, which could be used for real-time bioimaging and enhanced osseointegration. The PEEK-PCS surface exhibits viscoelastic properties, enabling it to conform to complex tissue geometries and effectively alleviate surface stress. Furthermore, PEEK-PCS modulates the inflammatory response by promoting macrophage M2 phenotypic polarization and reducing the expression of inflammatory factors. Additionally, PEEK-PCS promotes the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), significantly enhancing the osseointegration and osteogenesis ability of PEEK implants. Notably, PEEK-PCS demonstrates excellent autofluorescence properties both in vitro and in vivo, along with remarkable fluorescence stability over 14 d in vivo, suggesting real-time tracking potential of bioimaging. Compared to traditional coated implants, PEEK-PCS provides distinct advantages in surface adhesion, mechanical compatibility, real-time bioimaging, and osseointegration, representing a promising solution for implant-related bone repair.
Collapse
Affiliation(s)
- Hongyun Ma
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
| | - Jin Tong
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Xiaochen Su
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, P. R. China
| | - Liang Liu
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Jingqi Liang
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Jianbo Sun
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Jun Lu
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| | - Yingang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Bo Lei
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, P. R. China
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Hongmou Zhao
- Foot and Ankle Surgery Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, P. R. China
| |
Collapse
|
3
|
Xiaojie X, Jinbing C, Yiling C, JingJing S, Yuan L, Yu P, Hao Y, Hui C. A photo-thermal dual crosslinked chitosan-based hydrogel membrane for guided bone regeneration. Int J Biol Macromol 2025; 296:139712. [PMID: 39793779 DOI: 10.1016/j.ijbiomac.2025.139712] [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: 07/31/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Alveolar bone defects caused by inflammation or trauma jeopardize patients' oral functions. Guided bone regeneration (GBR) is widely used in repairing periodontal tissue, with barrier membranes play a crucial role in preserving the bone regeneration space. In this study, an injectable dual-crosslinked hydrogel was developed to improve the existing barrier membranes in flexibility and functionality. The hydrogel matrix, composed of methacrylated carboxymethyl chitosan (CMCS) reinforced with robust silk fibroin (SF), was further functionalized with bioactive glass (BG) particles to promote bone regeneration. The pre-gel solution achieved a fast-curing process under visible light and at body temperature. Further, the composite hydrogels presented good biocompatibility, biodegradability, resilience, alongside in vitro barrier effect against human gingival fibroblasts (HGFs). It significantly enhanced osteogenic differentiation and angiogenesis of bone marrow mesenchymal stem cells (BMSCs), facilitate the tube formation of human umbilical vein endothelial cells (HUVECs), and inhibit Staphylococcus aureus and Porphyromonas gingivalis. In a rat skull defect model, the osteogenic performance of hydrogels was comparable with that of collagen membranes (Bio-Gide®). Overall, this in-situ gel-forming barrier material served as a stable carrier for bioactive ions and a biomineralized scaffold for tissue ingrowth, supporting the enhancement of GBR technique.
Collapse
Affiliation(s)
- Xing Xiaojie
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| | - Chen Jinbing
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| | - Cheng Yiling
- Stomatological Hospital of Xiamen Medical College, Xiamen, Fujian 361008, China
| | - Su JingJing
- Stomatological Hospital of Xiamen Medical College, Xiamen, Fujian 361008, China
| | - Liu Yuan
- Institute of Stomatology & Research Center of Dental Esthetics and Biomechanics, School and Hospital of Stomatology, Fujian Medical University, 246 Yangqiao Zhong Road, Fuzhou, Fujian 350002, China
| | - Pan Yu
- Institute of Stomatology & Research Center of Dental Esthetics and Biomechanics, School and Hospital of Stomatology, Fujian Medical University, 246 Yangqiao Zhong Road, Fuzhou, Fujian 350002, China
| | - Yu Hao
- Institute of Stomatology & Research Center of Dental Esthetics and Biomechanics, School and Hospital of Stomatology, Fujian Medical University, 246 Yangqiao Zhong Road, Fuzhou, Fujian 350002, China.
| | - Cheng Hui
- Institute of Stomatology & Research Center of Dental Esthetics and Biomechanics, School and Hospital of Stomatology, Fujian Medical University, 246 Yangqiao Zhong Road, Fuzhou, Fujian 350002, China.
| |
Collapse
|
4
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
5
|
Shi Y, Wang Z, Xu W, Yu X, Gao B, Zhou X, Chen J, Jia K, Cheang LH, Tam MS, Wang H, Zheng X, Wu T. Preparation and osteogenesis of a multiple crosslinking silk fibroin/carboxymethyl chitosan/sodium alginate composite scaffold loading with mesoporous silica/poly (lactic acid-glycolic acid) microspheres. J Biomater Appl 2025; 39:578-591. [PMID: 39264258 DOI: 10.1177/08853282241281439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Large bone defect repair is a striking challenge in orthopedics. Currently, inorganic-organic composite scaffolds are considered as a promising approach to these bone regeneration. Silicon ions (Si4+) are bioactive and beneficial to bone regeneration and Si4+-containing inorganic mesoporous silica (MS) can effectively load drugs for bone repair. To better control the release of drug, we prepared biodegradable MS/PLGA (MP) microspheres. MP loaded organic silk fibroin/carboxymethyl chitosan/sodium alginate (MP/SF/CMCS/SA) composite scaffolds were further constructed by genipin and Ca2+ crosslinking. All MP/SF/CMCS/SA scaffolds had good swelling ability, degradation rate and high porosity. The incorporation of 1% MP significantly enhanced the compressive strength of composite scaffolds. Besides, MP loaded scaffold showed a sustained release of Si4+ and Ca2+. Moreover, the release rate of rhodamine (a model drug) of MP/SF/CMCS/SA scaffolds was obviously lower than that of MP. When culturing with rat bone marrow mesenchymal stem cells, scaffolds with 1% MP displayed good proliferation, adhesion and enhanced osteogenic differentiation ability. Based on the results above, the addition of 1% MP in SF/CMCS/SA scaffolds is a prospective way for drug release in bone regeneration and is promising for further in vivo bone repair applications.
Collapse
Affiliation(s)
- Yiwan Shi
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, PR China
| | - Zhaozhen Wang
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, PR China
| | - Weikang Xu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Xiaolu Yu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, PR China
| | - Botao Gao
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Xinting Zhou
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiwen Chen
- The Affiliated Shunde Hospital of Jinan University, The Second People's Hospital of Shunde, Foshan, Guangdong, China
| | - Kunfeng Jia
- Herbal Kingdom Pharmaceutical Co., Ltd, Macau, China
| | - Lek Hang Cheang
- Department of Orthopedic Surgery, Centro Hospitalar Conde de Sao Januario, Macau, China
| | - Man Seng Tam
- IAN WO Medical Center, Macao Special Administrative Region, People's Republic of China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, PR China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, PR China
| | - Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological And Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
6
|
Abdian N, Soltani Zangbar H, Etminanfar M, Hamishehkar H. 3D chitosan/hydroxyapatite scaffolds containing mesoporous SiO2-HA particles: A new step to healing bone defects. Int J Biol Macromol 2024; 278:135014. [PMID: 39181354 DOI: 10.1016/j.ijbiomac.2024.135014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Biocompatible scaffolds with high mechanical strengths that contain biodegradable components could boost bone regeneration compared with nondegradable bone repair materials. In this study, porous chitosan (CS)/hydroxyapatite (HA) scaffolds containing mesoporous SiO2-HA particles were fabricated through the freeze-drying process. According to field emission scanning electron microscopy (FESEM) results, combining mesoporous SiO2-HA particles in CS/HA scaffolds led to a uniform porous structure. It decreased pore sizes from 320 ± 1.1 μm to 145 ± 1.4 μm. Moreover, the compressive strength value of this scaffold was 25 ± 1.2 MPa. The in-vitro approaches exhibited good sarcoma osteogenic cell line (SAOS-2) adhesion, spreading, and proliferation, indicating that the scaffolds provided a suitable environment for cell cultivation. Also, in-vivo analyses in implanted defect sites of rats proved that the CS/HA/mesoporous SiO2-HA scaffolds could promote bone regeneration via enhancing osteoconduction and meliorating the expression of osteogenesis gene to 19.31 (about 5-fold higher compared to the control group) by exposing them to the bone-like precursors. Further, this scaffold's new bone formation percentage was equal to 90 % after 21 days post-surgery. Therefore, incorporating mesoporous SiO2-HA particles into CS/HA scaffolds can suggest a new future tissue engineering and regeneration strategy.
Collapse
Affiliation(s)
- Nesa Abdian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Advanced Materials, Faculty of Materials Engineering, Sahand University of Technology, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamadreza Etminanfar
- Research Center for Advanced Materials, Faculty of Materials Engineering, Sahand University of Technology, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Akoa DM, Hélary C, Foda A, Chaussain C, Poliard A, Coradin T. Silicon impacts collagen remodelling and mineralization by human dental pulp stem cells in 3D pulp-like matrices. Dent Mater 2024; 40:1390-1399. [PMID: 38908960 DOI: 10.1016/j.dental.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVES Silicon-releasing biomaterials are widely used in the field of dentistry. However, unlike bone, very little is known about the role of silicon on dental tissue formation and repair. This study investigates the influence of silicic acid on the survival, differentiation and mineralizing ability of human dental pulp stem cells (hDPSCs) in 3D pulp-like environments METHODS: Dense type I collagen hydrogels seeded with hDPSCs were cultured over 4 weeks in the presence of silicic acid at physiological (10 μM) and supraphysiological (100 μM) concentrations. Cell viability and proliferation were studied by Alamar Blue and live/dead staining. The collagen network was investigated using second harmonic generation imaging. Mineral deposition was monitored by histology and scanning electron microscopy. Gene expression of mineralization- and matrix remodeling-associated proteins was studied by qPCR. RESULTS Presence of silicic acid did not show any significant influence on cell survival, metabolic activity and gene expression of key mineralization-related proteins (ALP, OCN, BSP). However, it induced enhanced cell clustering and delayed expression of matrix remodeling-associated proteins (MMP13, Col I). OPN expression and mineral deposition were inhibited at 100 μM. It could be inferred that silicic acid has no direct cellular effect but rather interacts with the collagen network, leading to a modification of the cell-matrix interface. SIGNIFICANCE Our results offer advanced insights on the possible role of silicic acid, as released by pulp capping calcium silicates biomaterials, in reparative dentine formation. More globally, these results interrogate the possible role of Si in pulp pathophysiology.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Asmaa Foda
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Catherine Chaussain
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France; AP-HP Service de médecine bucco-dentaire, Hôpital Bretonneau, Paris, France
| | - Anne Poliard
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France.
| |
Collapse
|
8
|
Alashi S, Alkhouri I, Alghoraibi I, Kochaji N, Houri A, Karkoutly M. Evaluating various properties of nanohydroxyapatite synthesized from eggshells and dual-doped with Si 4+ and Zn 2+: An in vitro study. Heliyon 2024; 10:e35907. [PMID: 39224256 PMCID: PMC11366878 DOI: 10.1016/j.heliyon.2024.e35907] [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: 04/18/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study aimed to evaluate morphological, chemical and biocompatible properties of nanohydroxyapatite (N-HA) synthesized from eggshells and dual-doped with Si4+ and Zn2+. METHODS In the current study, N-HA was synthesized from chicken eggshells using the wet chemical precipitation method and doped with Si4+ and Zn2+. The physical assessment was carried out using field emission scanning electron microscopy (FE-SEM), energy dispersive X-ray (EDX) analysis, and X-ray diffraction (XRD) analysis. Crystal size was calculated using the Scherrer equation. Cytotoxicity was studied in vitro using the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) cytotoxicity assay. The optical density (OD) of each well was obtained and recorded at 570 nm for 24 h (t1), 48 h (t2), 72 h (t3), and 5 days (t4) using a microplate reader. RESULTS The results of Si-Zn-doped HA showed a high specific surface area with an irregular nano-sized spherical particle structure. The atomic percentage provided the ratio of calcium to phosphate; for non-doped HA, the atomic Ca/P ratio was 1.6, but for Si-Zn-doped HA, where Zn+2 Ca and Si + replaced 4 substituted P, the atomic ratio (Ca + Zn)/(P + Si) was 1.76. The average crystal size of Si-Zn-doped HA was 46 nm, while for non-doped HA it was 61 nm. both samples were non-toxic and statistically significantly less viable than the control group After 5 days, the mean cell viability of Si-Zn-doped HA (79.17 ± 2.18) was higher than that of non-doped HA (76.26 ± 1.71) (P = 0.091). CONCLUSIONS The MTT assay results showed that Si-Zn-doped HA is biocompatible. In addition, it showed characteristic physiochemical properties of a large surface area with interconnected porosity.
Collapse
Affiliation(s)
- Shaza Alashi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Isam Alkhouri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Ibrahim Alghoraibi
- Department of Physics, Faculty of Science, Damascus University, Damascus, Syrian Arab Republic
| | - Nabil Kochaji
- Department of Oral Pathology, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| | - Abdullah Houri
- Department of Physics, Faculty of Science, Damascus University, Damascus, Syrian Arab Republic
| | - Mawia Karkoutly
- Department of Pediatric Dentistry, Faculty of Dentistry, Damascus University, Damascus, Syrian Arab Republic
| |
Collapse
|
9
|
Tong Y, Yuan J, Li Z, Deng C, Cheng Y. Drug-Loaded Bioscaffolds for Osteochondral Regeneration. Pharmaceutics 2024; 16:1095. [PMID: 39204440 PMCID: PMC11360256 DOI: 10.3390/pharmaceutics16081095] [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: 05/12/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Osteochondral defect is a complex tissue loss disease caused by arthritis, high-energy trauma, and many other reasons. Due to the unique structural characteristics of osteochondral tissue, the repair process is sophisticated and involves the regeneration of both hyaline cartilage and subchondral bone. However, the current clinical treatments often fall short of achieving the desired outcomes. Tissue engineering bioscaffolds, especially those created via three-dimensional (3D) printing, offer promising solutions for osteochondral defects due to their precisely controllable 3D structures. The microstructure of 3D-printed bioscaffolds provides an excellent physical environment for cell adhesion and proliferation, as well as nutrient transport. Traditional 3D-printed bioscaffolds offer mere physical stimulation, while drug-loaded 3D bioscaffolds accelerate the tissue repair process by synergistically combining drug therapy with physical stimulation. In this review, the physiological characteristics of osteochondral tissue and current treatments of osteochondral defect were reviewed. Subsequently, the latest progress in drug-loaded bioscaffolds was discussed and highlighted in terms of classification, characteristics, and applications. The perspectives of scaffold design, drug control release, and biosafety were also discussed. We hope this article will serve as a valuable reference for the design and development of osteochondral regenerative bioscaffolds and pave the way for the use of drug-loaded bioscaffolds in clinical therapy.
Collapse
Affiliation(s)
| | | | | | - Cuijun Deng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (Y.T.); (J.Y.); (Z.L.)
| | - Yu Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, China; (Y.T.); (J.Y.); (Z.L.)
| |
Collapse
|
10
|
Shrestha S, Tieu T, Wojnilowicz M, Voelcker NH, Forsythe JS, Frith JE. Delivery of miRNAs Using Porous Silicon Nanoparticles Incorporated into 3D Hydrogels Enhances MSC Osteogenesis by Modulation of Fatty Acid Signaling and Silicon Degradation. Adv Healthc Mater 2024; 13:e2400171. [PMID: 38657207 DOI: 10.1002/adhm.202400171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Strategies incorporating mesenchymal stromal cells (MSC), hydrogels and osteoinductive signals offer promise for bone repair. Osteoinductive signals such as growth factors face challenges in clinical translation due to their high cost, low stability and immunogenicity leading to interest in microRNAs as a simple, inexpensive and powerful alternative. The selection of appropriate miRNA candidates and their efficient delivery must be optimised to make this a reality. This study evaluated pro-osteogenic miRNAs and used porous silicon nanoparticles modified with polyamidoamine dendrimers (PAMAM-pSiNP) to deliver these to MSC encapsulated within gelatin-PEG hydrogels. miR-29b-3p, miR-101-3p and miR-125b-5p are strongly pro-osteogenic and are shown to target FASN and ELOVL4 in the fatty acid biosynthesis pathway to modulate MSC osteogenesis. Hydrogel delivery of miRNA:PAMAM-pSiNP complexes enhanced transfection compared to 2D. The osteogenic potential of hBMSC in hydrogels with miR125b:PAMAM-pSiNP complexes is evaluated. Importantly, a dual-effect on osteogenesis occurred, with miRNAs increasing expression of alkaline phosphatase (ALP) and Runt-related transcription factor 2 (RUNX2) whilst the pSiNPs enhanced mineralisation, likely via degradation into silicic acid. Overall, this work presents insights into the role of miRNAs and fatty acid signalling in osteogenesis, providing future targets to improve bone formation and a promising system to enhance bone tissue engineering.
Collapse
Affiliation(s)
- Surakshya Shrestha
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Terence Tieu
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Marcin Wojnilowicz
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
11
|
Olăreț E, Dinescu S, Dobranici AE, Ginghină RE, Voicu G, Mihăilescu M, Curti F, Banciu DD, Sava B, Amarie S, Lungu A, Stancu IC, Mastalier BSM. Osteoblast responsive biosilica-enriched gelatin microfibrillar microenvironments. BIOMATERIALS ADVANCES 2024; 161:213894. [PMID: 38796956 DOI: 10.1016/j.bioadv.2024.213894] [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: 10/26/2023] [Revised: 02/09/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Engineering of scaffolds for bone regeneration is often inspired by the native extracellular matrix mimicking its composite fibrous structure. In the present study, we used low loadings of diatomite earth (DE) biosilica to improve the bone regeneration potential of gelatin electrospun fibrillar microenvironments. We explored the effect of increasing the DE content from 1 % to 3 % and 5 %, respectively, on the physico-chemical properties of the fibrous scaffolds denoted FG_DE1, FG_DE3, FG_DE5, regarding the aqueous media affinity, stability under simulated physiological conditions, morphology characteristics, and local mechanical properties at the surface. The presence of biosilica generated composite structures with lower swelling degrees and higher stiffness when compared to gelatin fibers. Increasing DE content led to higher Young modulus, while the stability of the protein matrix in PBS, at 37 °C, over 21 was significantly decreased by the presence of diatomite loadings. The best preosteoblast response was obtained for FG_DE3, with enhanced mineralization during the osteogenic differentiation when compared to the control sample without diatomite. 5 % DE in FG_DE5 proved to negatively influence cells' metabolic activity and morphology. Hence, the obtained composite microfibrillar scaffolds might find application as osteoblast-responsive materials for bone tissue engineering.
Collapse
Affiliation(s)
- Elena Olăreț
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; Research Institute of the University of Bucharest (ICUB), 050663 Bucharest, Romania
| | - Alexandra-Elena Dobranici
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Raluca-Elena Ginghină
- Research and Innovation Center for CBRN Defense and Ecology, 041327 Bucharest, Romania
| | - Georgeta Voicu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Mona Mihăilescu
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; Faculty of Applied Sciences, National University of Science and Technology Politehnica Bucharest, 060042 Bucharest, Romania
| | - Filis Curti
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; Zentiva SA, 50, Theodor Pallady, 032266 Bucharest, Romania
| | - Daniel Dumitru Banciu
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | | | | | - Adriana Lungu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Izabela-Cristina Stancu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania.
| | - Bogdan Stelian Manolescu Mastalier
- University of Medicine and Pharmacy Carol Davila, Bucharest, Romania; Department of General Surgery, Colentina Clinical Hospital, 072202 Bucharest, Romania
| |
Collapse
|
12
|
Lange M, Babczyk P, Tobiasch E. Exosomes: A New Hope for Angiogenesis-Mediated Bone Regeneration. Int J Mol Sci 2024; 25:5204. [PMID: 38791243 PMCID: PMC11120942 DOI: 10.3390/ijms25105204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Bone is a metabolically dynamic structure that is generally remodeled throughout the lifetime of an individual but often causes problems with increasing age. A key player for bone development and homeostasis, but also under pathological conditions, is the bone vasculature. This complex system of arteries, veins, and capillaries forms distinct structures where each subset of endothelial cells has important functions. Starting with the basic process of angiogenesis and bone-specific blood vessel formation, coupled with initial bone formation, the importance of different vascular structures is highlighted with respect to how these structures are maintained or changed during homeostasis, aging, and pathological conditions. After exemplifying the current knowledge on bone vasculature, this review will move on to exosomes, a novel hotspot of scientific research. Exosomes will be introduced starting from their discovery via current isolation procedures and state-of-the-art characterization to their role in bone vascular development, homeostasis, and bone regeneration and repair while summarizing the underlying signal transduction pathways. With respect to their role in these processes, especially mesenchymal stem cell-derived extracellular vesicles are of interest, which leads to a discussion on patented applications and an update on ongoing clinical trials. Taken together, this review provides an overview of bone vasculature and bone regeneration, with a major focus on how exosomes influence this intricate system, as they might be useful for therapeutic purposes in the near future.
Collapse
Affiliation(s)
- Martin Lange
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Patrick Babczyk
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| |
Collapse
|
13
|
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
|
14
|
Dahiya A, Chaudhari VS, Kushram P, Bose S. 3D Printed SiO 2-Tricalcium Phosphate Scaffolds Loaded with Carvacrol Nanoparticles for Bone Tissue Engineering Application. J Med Chem 2024; 67:2745-2757. [PMID: 38146876 PMCID: PMC11164277 DOI: 10.1021/acs.jmedchem.3c01884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Bone damage resulting from trauma or aging poses challenges in clinical settings that need to be addressed using bone tissue engineering (BTE). Carvacrol (CA) possesses anti-inflammatory, anticancer, and antibacterial properties. Limited solubility and physicochemical stability restrict its biological activity, requiring a stable carrier system for delivery. Here, we investigate the utilization of a three-dimensional printed (3DP) SiO2-doped tricalcium phosphate (TCP) scaffold functionalized with carvacrol-loaded lipid nanoparticles (CA-LNPs) to improve bone health. It exhibits a negative surface charge with an entrapment efficiency of ∼97% and size ∼129 nm with polydispersity index (PDI) and zeta potential values of 0.18 and -16 mV, respectively. CA-LNPs exhibit higher and long-term release over 35 days. The CA-LNP loaded SiO2-doped TCP scaffold demonstrates improved antibacterial properties against Staphylococcus aureus and Pseudomonas aeruginosa by >90% reduction in bacterial growth. Functionalized scaffolds result in 3-fold decrease and 2-fold increase in osteosarcoma and osteoblast cell viability, respectively. These findings highlight the therapeutic potential of the CA-LNP loaded SiO2-doped TCP scaffold for bone defect treatment.
Collapse
Affiliation(s)
- Aditi Dahiya
- Department of Chemistry, Washington State University, Pullman, Washington 99164, United States
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Vishal Sharad Chaudhari
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Priya Kushram
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Susmita Bose
- Department of Chemistry, Washington State University, Pullman, Washington 99164, United States
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
15
|
Aghajanzadeh MS, Imani R, Nazarpak MH, McInnes SJP. Augmented physical, mechanical, and cellular responsiveness of gelatin-aldehyde modified xanthan hydrogel through incorporation of silicon nanoparticles for bone tissue engineering. Int J Biol Macromol 2024; 259:129231. [PMID: 38185310 DOI: 10.1016/j.ijbiomac.2024.129231] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
Bioactive scaffolds fabricated from a combination of organic and inorganic biomaterials are a promising approach for addressing defects in bone tissue engineering. In the present study, a self-crosslinked nanocomposite hydrogel, composed of gelatin/aldehyde-modified xanthan (Gel-AXG) is successfully developed by varying concentrations of porous silicon nanoparticles (PSiNPs). The effect of PSiNPs incorporation on physical, mechanical, and biological performance of the nanocomposite hydrogel is evaluated. Morphological analysis reveals formation of highly porous 3D microstructures with interconnected pores in all nanocomposite hydrogels. Increased content of PSiNPs results in a lower swelling ratio, reduced porosity and pore size, which in turn impeded media penetration and slowed down the degradation process. In addition, remarkable enhancements in dynamic mechanical properties are observed in Gel-AXG-8%Si (compressive strength: 0.6223 MPa at 90 % strain and compressive modulus: 0.054 MPa), along with improved biomineralization ability via hydroxyapatite formation after immersion in simulated body fluid (SBF). This optimized nanocomposite hydrogel provides a sustained release of Si ions at safe dose levels. Furthermore, in-vitro cytocompatibility studies using MG-63 cells exhibited remarkable performance in terms of cell attachment, proliferation, and ALP activity for Gel-AXG-8%Si. These findings suggest that the prepared nanocomposite hydrogel holds promising potential as a scaffold for bone tissue engineering.
Collapse
Affiliation(s)
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Masoumeh Haghbin Nazarpak
- New Technologies Research Center, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Steven J P McInnes
- UniSA STEM, Mawson Lakes Campus, University of South Australia, Mawson Lakes, South Australia, Australia
| |
Collapse
|
16
|
Han Z, Xiong J, Jin X, Dai Q, Han M, Wu H, Yang J, Tang H, He L. Advances in reparative materials for infectious bone defects and their applications in maxillofacial regions. J Mater Chem B 2024; 12:842-871. [PMID: 38173410 DOI: 10.1039/d3tb02069j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Infectious bone defects are characterized by the partial loss or destruction of bone tissue resulting from bacterial contaminations subsequent to diseases or external injuries. Traditional bone transplantation and clinical methods are insufficient in meeting the treatment demands for such diseases. As a result, researchers have increasingly focused on the development of more sophisticated biomaterials for improved therapeutic outcomes in recent years. This review endeavors to investigate specific reparative materials utilized for the treatment of infectious bone defects, particularly those present in the maxillofacial region, with a focus on biomaterials capable of releasing therapeutic substances, functional contact biomaterials, and novel physical therapy materials. These biomaterials operate via heightened antibacterial or osteogenic properties in order to eliminate bacteria and/or stimulate bone cells regeneration in the defect, ultimately fostering the reconstitution of maxillofacial bone tissue. Based upon some successful applications of new concept materials in bone repair of other parts, we also explore their future prospects and potential uses in maxillofacial bone repair later in this review. We highlight that the exploration of advanced biomaterials holds promise in establishing a solid foundation for the development of more biocompatible, effective, and personalized treatments for reconstructing infectious maxillofacial defects.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jingdi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xiaohan Jin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qinyue Dai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Mingyue Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Hongkun Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Haiqin Tang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Libang He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
De Mori A, Heyraud A, Tallia F, Blunn G, Jones JR, Roncada T, Cobb J, Al-Jabri T. Ovine Mesenchymal Stem Cell Chondrogenesis on a Novel 3D-Printed Hybrid Scaffold In Vitro. Bioengineering (Basel) 2024; 11:112. [PMID: 38391598 PMCID: PMC10886199 DOI: 10.3390/bioengineering11020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
This study evaluated the use of silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) 3D-printed scaffolds, with channel sizes of either 200 (SC-200) or 500 (SC-500) µm, as biomaterials to support the chondrogenesis of sheep bone marrow stem cells (oBMSC), under in vitro conditions. The objective was to validate the potential use of SiO2/PTHF/PCL-diCOOH for prospective in vivo ovine studies. The behaviour of oBMSC, with and without the use of exogenous growth factors, on SiO2/PTHF/PCL-diCOOH scaffolds was investigated by analysing cell attachment, viability, proliferation, morphology, expression of chondrogenic genes (RT-qPCR), deposition of aggrecan, collagen II, and collagen I (immunohistochemistry), and quantification of sulphated glycosaminoglycans (GAGs). The results showed that all the scaffolds supported cell attachment and proliferation with upregulation of chondrogenic markers and the deposition of a cartilage extracellular matrix (collagen II and aggrecan). Notably, SC-200 showed superior performance in terms of cartilage gene expression. These findings demonstrated that SiO2/PTHF/PCL-diCOOH with 200 µm pore size are optimal for promoting chondrogenic differentiation of oBMSC, even without the use of growth factors.
Collapse
Affiliation(s)
- Arianna De Mori
- School of Pharmacy and Biomedical Science, University of Portsmouth, St Micheal's Building, White Swan Road, Portsmouth PO1 2DT, UK
| | - Agathe Heyraud
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Francesca Tallia
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Science, University of Portsmouth, St Micheal's Building, White Swan Road, Portsmouth PO1 2DT, UK
| | - Julian R Jones
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Tosca Roncada
- Trinity Center for Biomedical Engineering, Trinity Biomedical Science Institute, Trinity College Dublin, 152-160 Pearse Street, DO2 R590 Dublin, Ireland
| | - Justin Cobb
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Talal Al-Jabri
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
18
|
Shimizu K, Nishi M, Sakate Y, Kawanami H, Bito T, Arima J, Leria L, Maldonado M. Silica-associated proteins from hexactinellid sponges support an alternative evolutionary scenario for biomineralization in Porifera. Nat Commun 2024; 15:181. [PMID: 38185711 PMCID: PMC10772126 DOI: 10.1038/s41467-023-44226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Metazoans use silicon traces but rarely develop extensive silica skeletons, except for the early-diverging lineage of sponges. The mechanisms underlying metazoan silicification remain incompletely understood, despite significant biotechnological and evolutionary implications. Here, the characterization of two proteins identified from hexactinellid sponge silica, hexaxilin and perisilin, supports that the three classes of siliceous sponges (Hexactinellida, Demospongiae, and Homoscleromorpha) use independent protein machineries to build their skeletons, which become non-homologous structures. Hexaxilin forms the axial filament to intracellularly pattern the main symmetry of the skeletal parts, while perisilin appears to operate in their thickening, guiding extracellular deposition of peripheral silica, as does glassin, a previously characterized hexactinellid silicifying protein. Distant hexaxilin homologs occur in some bilaterians with siliceous parts, suggesting putative conserved silicifying activity along metazoan evolution. The findings also support that ancestral Porifera were non-skeletonized, acquiring silica skeletons only after diverging into major classes, what reconciles molecular-clock dating and the fossil record.
Collapse
Affiliation(s)
- Katsuhiko Shimizu
- Platform for Community-based Research and Education, Tottori University, 4-101, Koyama-cho, Minami, Tottori, 680-8550, Japan.
| | - Michika Nishi
- Division of Agricultural Science, Graduate studies of Sustainability Science, Tottori University Graduate School, 4-101, Koyama-cho, Minami, Tottori, 680-8553, Japan
| | - Yuto Sakate
- Division of Agricultural Science, Graduate studies of Sustainability Science, Tottori University Graduate School, 4-101, Koyama-cho, Minami, Tottori, 680-8553, Japan
| | - Haruka Kawanami
- Department of Life Environmental Agriculture, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho, Minami, Tottori, 680-8553, Japan
| | - Tomohiro Bito
- Department of Life Environmental Agriculture, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho, Minami, Tottori, 680-8553, Japan
| | - Jiro Arima
- Department of Life Environmental Agriculture, Faculty of Agriculture, Tottori University, 4-101, Koyama-cho, Minami, Tottori, 680-8553, Japan
| | - Laia Leria
- Sponge Ecobiology and Biotechnology Group, Center for Advanced Studies of Blanes (CEAB, CSIC), Blanes, 17300, Spain
| | - Manuel Maldonado
- Sponge Ecobiology and Biotechnology Group, Center for Advanced Studies of Blanes (CEAB, CSIC), Blanes, 17300, Spain.
| |
Collapse
|
19
|
Carnieri MV, Garcia DDF, Voltolini R, Volpato N, Mafra M, Bernardelli EA, Stimamiglio MA, Rebelatto CK, Correa A, Berti LF, Marcon BH. Cytocompatible and osteoconductive silicon oxycarbide glass scaffolds 3D printed by DLP: a potential material for bone tissue regeneration. Front Bioeng Biotechnol 2024; 11:1297327. [PMID: 38239914 PMCID: PMC10794595 DOI: 10.3389/fbioe.2023.1297327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Bone lesions affect individuals of different age groups, compromising their daily activities and potentially leading to prolonged morbidity. Over the years, new compositions and manufacturing technologies were developed to offer customized solutions to replace injured tissue and stimulate tissue regeneration. This work used digital light processing (DPL) technology for three-dimensional (3D) printing of porous structures using pre-ceramic polymer, followed by pyrolysis to obtain SiOC vitreous scaffolds. The SiOC scaffolds produced had an amorphous structure (compatible with glass) with an average porosity of 72.69% ± 0.99, an average hardness of 935.1 ± 71.0 HV, and an average maximum flexural stress of 7.8 ± 1.0 MPa, similar to cancellous bone tissue. The scaffolds were not cytotoxic and allowed adult stem cell adhesion, growth, and expansion. After treatment with osteoinductive medium, adult stem cells in the SiOC scaffolds differentiated to osteoblasts, assuming a tissue-like structure, with organization in multiple layers and production of a dense fibrous matrix rich in hydroxyapatite. The in vitro analyses supported the hypothesis that the SiOC scaffolds produced in this work were suitable for use as a bone substitute for treating critically sized lesions, with the potential to stimulate the gradual process of regeneration of the native tissue. The data obtained stimulate the continuity of studies with the SiOC scaffolds developed in this work, paving the way for evaluating safety and biological activity in vivo.
Collapse
Affiliation(s)
- Matheus Versão Carnieri
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Daniele de Freitas Garcia
- Laboratory of Basic Biology of Stem Cells (LABCET), Carlos Chagas Institute—FIOCRUZ-PR, Curitiba, Brazil
| | - Rafael Voltolini
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Neri Volpato
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Marcio Mafra
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Euclides Alexandre Bernardelli
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Marco Augusto Stimamiglio
- Laboratory of Basic Biology of Stem Cells (LABCET), Carlos Chagas Institute—FIOCRUZ-PR, Curitiba, Brazil
| | | | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells (LABCET), Carlos Chagas Institute—FIOCRUZ-PR, Curitiba, Brazil
| | - Lucas Freitas Berti
- Department of Mechanical Engineering, Postgraduate Program in Mechanical and Materials Engineering, Universidade Tecnológica Federal Do Parana, Curitiba, Brazil
| | - Bruna Hilzendeger Marcon
- Laboratory of Basic Biology of Stem Cells (LABCET), Carlos Chagas Institute—FIOCRUZ-PR, Curitiba, Brazil
- Confocal and Eletronic Microscopy Facility (RPT07C), Carlos Chagas Institute—FIOCRUZ-PR, Curitiba, Brazil
| |
Collapse
|
20
|
Wee CY, Lim QRT, Xu X, Yang Z, Wang D, Thian ES. Characterization and in-vitro assessment of silicon-based apatite microspheres for bone tissue engineering applications. J Biomed Mater Res B Appl Biomater 2024; 112:e35349. [PMID: 38247239 DOI: 10.1002/jbm.b.35349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/15/2023] [Accepted: 10/30/2023] [Indexed: 01/23/2024]
Abstract
In the field of bone tissue engineering, silicon (Si) has been found as an essential element for bone growth. However, the use of silicon in bioceramics microspheres remains limited. In this work, different weight percentages (0.8, 1.6, and 2.4 wt %) of silicon was incorporated into hydroxyapatite and fabricated into microspheres. 2.4 wt % of Si incorporated into HAp microspheres (2.4 SiHAp) were found to enhance functional properties of the microspheres which resulted in improved cell viability of human mesenchymal stem cells (hMSCs), demonstrating rapid cell proliferation rates resulting in high cell density accumulated on the surface of the microspheres which in turn permitted better hMSCs differentiation into osteoblasts when validated by bone marker assays (Type I collagen, alkaline phosphatase, osteocalcin, and osteopontin) compared to apatite microspheres of lower wt % of Si incorporated and non-substituted HAp (2.4 SiHAp >1.6 SiHAp >0.8 SiHAp > HAp). SEM images displayed the densest cell population on 2.4 SiHAp surfaces with the greatest degree of cell stretching and bridging between neighboring microspheres. Incorporation of silicon into apatite microspheres was found to accelerate the rate and number of apatite nucleation sites formed when subjected to physiological conditions improving the interface between the microsphere scaffolds and bone forming cells, facilitating better adhesion and proliferation.
Collapse
Affiliation(s)
- Chien Yi Wee
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Quentin Ray Tjieh Lim
- Department of Material Science and Engineering, National University of Singapore, Singapore, Singapore
| | - Xin Xu
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Zhijie Yang
- Zhejiang Biocare Biotechnology Co. Ltd, Shaoxing, China
| | - Dong Wang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Eng San Thian
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Zhang D, Liu P. Biosynthesis of metal nanoparticles: Bioreduction and biomineralization. NANOTECHNOLOGY REVIEWS 2023; 12. [DOI: 10.1515/ntrev-2023-0170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Abstract
The biosynthesis of metal nanoparticles by plants, bacteria, and cells has been receiving considerable attention in recent years. The traditional synthesis of metal nanoparticles always needed high temperatures, high pressure, and toxic agents. However, the biosynthesis process (including bioreduction and biomineralization) is simpler, safe, economical, and green. The process of biosynthesis can insulate toxic agents, streamline flux, increase the transition efficiency of interactants, and improve the product yield. The biosynthesized metal nanoparticles share similar characteristics with traditional ones, serving as photosensors to achieve light-to-heat/energy transduction, or a drug delivery system. The biosynthetic metal nanoparticles thus could be widely applied in the medical field for disease diagnosis and treatment. It contributed a novel modality for the facile and green synthesis of metal nanoparticles. Increasing studies have been exploring the mechanism for the biosynthesis of metal nanoparticles, devoted to a controllable biosynthesis process. Combined with our previous studies on the biosynthesis of gold nanoparticles with green tea, tumor cells, and cell components, we reviewed the green methods of bioreduction and biomineralization of metal nanoparticles including the internal mechanism, aimed to make a comprehensive introduction to the biosynthesis of metal nanoparticles and relevant biomedical applications, and inspired further research.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Pharmacy, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Pengran Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , 430022 , China
| |
Collapse
|
22
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
23
|
Pańtak P, Czechowska JP, Zima A. The influence of silane coupling agents on the properties of α-TCP-based ceramic bone substitutes for orthopaedic applications. RSC Adv 2023; 13:34020-34031. [PMID: 38020001 PMCID: PMC10663883 DOI: 10.1039/d3ra06027f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/11/2023] [Indexed: 12/01/2023] Open
Abstract
Biomaterials based on α-TCP are highly recommended for medical applications due to their ability to bond chemically with bone tissue. However, in order to improve their physicochemical properties, modifications are needed. In this work, novel, hybrid α-TCP-based bone cements were developed and examinated. The influence of two different silane coupling agents (SCAs) - tetraethoxysilane (TEOS) and 3-glycidoxypropyl trimethoxysilane (GPTMS) on the properties of the final materials was investigated. Application of modifiers allowed us to obtain hybrid materials due to the presence of different bonds in their structure, for example between calcium phosphates and SCA molecules. The use of SCAs increased the compressive strength of the bone cements from 7.24 ± 0.35 MPa to 12.17 ± 0.48 MPa. Moreover, modification impacted the final setting time of the cements, reducing it from 11.0 to 6.5 minutes. The developed materials displayed bioactive potential in simulated body fluid. Presented findings demonstrate the beneficial influence of silane coupling agents on the properties of calcium phosphate-based bone substitutes and pave the way for their further in vitro and in vivo studies.
Collapse
Affiliation(s)
- Piotr Pańtak
- Faculty of Materials Science and Ceramics, AGH University of Science and Technology Mickiewicza Av. 30 30-058 Kraków Poland
| | - Joanna P Czechowska
- Faculty of Materials Science and Ceramics, AGH University of Science and Technology Mickiewicza Av. 30 30-058 Kraków Poland
| | - Aneta Zima
- Faculty of Materials Science and Ceramics, AGH University of Science and Technology Mickiewicza Av. 30 30-058 Kraków Poland
| |
Collapse
|
24
|
Gao S, Li J, Lei Q, Chen Y, Huang H, Yan F, Xiao L, Zhang T, Wang L, Wei R, Hu C. Calcium sulfate-Cu 2+ delivery system improves 3D-Printed calcium silicate artificial bone to repair large bone defects. Front Bioeng Biotechnol 2023; 11:1224557. [PMID: 37954016 PMCID: PMC10634439 DOI: 10.3389/fbioe.2023.1224557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/25/2023] [Indexed: 11/14/2023] Open
Abstract
There are still limitations in artificial bone materials used in clinical practice, such as difficulty in repairing large bone defects, the mismatch between the degradation rate and tissue growth, difficulty in vascularization, an inability to address bone defects of various shapes, and risk of infection. To solve these problems, our group designed stereolithography (SLA) 3D-printed calcium silicate artificial bone improved by a calcium sulfate-Cu2+ delivery system. SLA technology endows the scaffold with a three-dimensional tunnel structure to induce cell migration to the center of the bone defect. The calcium sulfate-Cu2+ delivery system was introduced to enhance the osteogenic activity of calcium silicate. Rapid degradation of calcium sulfate (CS) induces early osteogenesis in the three-dimensional tunnel structure. Calcium silicate (CSi) which degrades slowly provides mechanical support and promotes bone formation in bone defect sites for a long time. The gradient degradation of these two components is perfectly matched to the rate of repair in large bone defects. On the other hand, the calcium sulfate delivery system can regularly release Cu2+ in the temporal and spatial dimensions, exerting a long-lasting antimicrobial effect and promoting vascular growth. This powerful 3D-printed calcium silicate artificial bone which has rich osteogenic activity is a promising material for treating large bone defects and has excellent potential for clinical application.
Collapse
Affiliation(s)
- Shijie Gao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiawen Li
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingjian Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yan Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Feifei Yan
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tie Zhang
- Wuhan QISIDA Technology Development Co., Ltd., Wuhan, Hubei, China
| | - Linlong Wang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Renxiong Wei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao Hu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
25
|
Skwira A, Szewczyk A, Barros J, Laranjeira M, Monteiro FJ, Sądej R, Prokopowicz M. Biocompatible antibiotic-loaded mesoporous silica/bioglass/collagen-based scaffolds as bone drug delivery systems. Int J Pharm 2023; 645:123408. [PMID: 37703959 DOI: 10.1016/j.ijpharm.2023.123408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/28/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Local delivery of antibiotics has gained increasing interest in the treatment of osteomyelitis due to its effectiveness and safety. Since the regeneration of bone tissue at the site of infection is as important as bacterial eradication, implantable drug delivery systems should not only release the drugs in a proper manner but also exert the osseointegration capability. Herein, we present an implantable drug delivery system in a scaffold form with a unique set of features for local treatment of osteomyelitis. For the first time, collagen type I, ciprofloxacin-loaded mesoporous silica, and bioglass were combined to obtain scaffolds using the molding method. Drug-loaded mesoporous silica was blended with polydimethylsiloxane to prolong the drug release, whereas bioglass served as a remineralization agent. Collagen-silica scaffolds were evaluated in terms of physicochemical properties, drug release rate, mineralization potential, osteoblast response in vitro, antimicrobial activity, and biological properties using an in vivo preclinical model - chick embryo chorioallantoic membrane (CAM). The desirable multifunctionality of the proposed collagen-silica scaffolds was confirmed. They released the ciprofloxacin for 80 days, prevented biofilm development, and induced hydroxyapatite formation. Moreover, the resulting macroporous structure of the scaffolds promoted osteoblast attachment, infiltration, and proliferation. Collagen-silica scaffolds were also biocompatible and effectively integrated with CAM.
Collapse
Affiliation(s)
- Adrianna Skwira
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland; Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Adrian Szewczyk
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland.
| | - Joana Barros
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FEUP-Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, Rua Dr. Roberto Frias, s/n 4200-465, Porto, Portugal.
| | - Marta Laranjeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| | - Fernando Jorge Monteiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; FEUP-Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, Rua Dr. Roberto Frias, s/n 4200-465, Porto, Portugal.
| | - Rafał Sądej
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Magdalena Prokopowicz
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland.
| |
Collapse
|
26
|
Sivolella S, Brunello G, Nika E, Badocco D, Pastore P, Carturan SM, Bernardo E, Elsayed H, Biasetto L, Brun P. In vitro evaluation of granules obtained from 3D sphene scaffolds and bovine bone grafts: chemical and biological assays. J Mater Chem B 2023; 11:8775-8787. [PMID: 37665632 DOI: 10.1039/d3tb00499f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Sphene is an innovative bone graft material. The aim of this study was to investigate and compare the physicochemical and biological properties of Bio-Oss® (BO) and in-lab synthesized and processed sphene granules. BO granules of 1000-2000 μm (BO-L), 250-1000 μm (BO-S) and 100-200 μm (BO-p) for derived granules, and corresponding groups of sphene granules obtained from 3D printed blocks (SB-L, SB-S, SB-p) and foams (SF-L, SF-S and SF-p) were investigated. The following analyses were conducted: morphological analysis, specific surface area and porosity, inductively coupled plasma mass spectrometry (ICP-MS), cytotoxicity assay, Alizarin staining, bone-related gene expression, osteoblast migration and proliferation assays. All pulverized granules exhibited a similar morphology and SF-S resembled natural bone. Sphene-derived granules showed absence of micro- and mesopores and a low specific surface area. ICP-MS revealed a tendency for absorption of Ca and P for all BO samples, while sphene granules demonstrated a release of Ca. No cellular cytotoxicity was detected and osteoblastic phenotype in primary cells was observed, with significantly increased values for SF-L, SF-S, BO-L and BO-p. Further investigations are needed before clinical use can be considered.
Collapse
Affiliation(s)
- Stefano Sivolella
- Department of Neuroscience, Dentistry Section, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Giulia Brunello
- Department of Neuroscience, Dentistry Section, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
- Department of Oral Surgery, Universitätsklinikum Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Ervin Nika
- Department of Neuroscience, Dentistry Section, University of Padova, Via Giustiniani 2, 35128 Padova, Italy.
| | - Denis Badocco
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy.
| | - Paolo Pastore
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy.
| | - Sara M Carturan
- INFN-Laboratori Nazionali di Legnaro, Viale dell'Università 2, 35020, Legnaro, PD, Italy.
- Dipartimento di Fisica e Astronomia, Università di Padova, Via Marzolo 8, 5131, Padua, Italy
| | - Enrico Bernardo
- Department of Industrial Engineering, University of Padova, Via F. Marzolo 9, 35131 Padova, Italy.
| | - Hamada Elsayed
- Department of Industrial Engineering, University of Padova, Via F. Marzolo 9, 35131 Padova, Italy.
- Refractories, Ceramics and Building Materials Department, National Research Centre, El Buhouth Str., Cairo 12622, Egypt
| | - Lisa Biasetto
- Department of Management and Engineering, University of Padova, Stradella San Nicola 3, 36100 Vicenza, Italy.
| | - Paola Brun
- Department of Molecular Medicine, Section of Microbiology, University of Padova, via A. Gabelli, 63, 35121 Padova, Italy.
| |
Collapse
|
27
|
Li X, Yang B, Xu M, Li F, Geng Z, Cui W, Sun X, Li Y, Liu Y. Doped Multiple Nanoparticles with Hydroxyapatite Coating Show Diverse Health Effects in vivo. Int J Nanomedicine 2023; 18:5031-5054. [PMID: 37701820 PMCID: PMC10493156 DOI: 10.2147/ijn.s417929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction The lack of osteoinductive, angiogenic and antimicrobial properties of hydroxyapatite coatings (HA) on titanium surfaces severely limits their use in orthopedic and dental implants. Therefore, we doped SiO2, Gd2O3 and CeO2 nanoparticles into HA to fabricate a HASiGdCe coating with a combination of decent antibacterial, angiogenic and osteogenic properties by the plasma spraying technique. Methods The HASiGdCe coating was analyzed by SEM (EDS), surface roughness tests, contact angle tests, XRD, FTIR spectroscopy, tensile tests and electrochemical dynamic polarization tests. Methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa (PAO-1) were used as representative bacteria to verify the antibacterial properties of the HASiGdCe coating. We evaluated the cytocompatibility and in vitro osteoinductivity of the HASiGdCe coating by investigating its effect on the cell viability and osteogenic differentiation of MC3T3-E1 cells. We assessed the in vitro angiogenic activity of the HASiGdCe coating by migration assay, tube formation assay, and RT‒PCR analysis of angiogenic genes in HUVECs. Finally, we used infected animal femur models to investigate the biosafety, antimicrobial and osteointegration properties of the HASiGdCe coating in vivo. Results Through various characterization experiments, we demonstrated that the HASiGdCe coating has suitable microscopic morphology, physical phase characteristics, bonding strength and bioactivity to meet the coating criteria for orthopedic implants. The HASiGdCe coating can release Gd3+ and Ce4+, showing strong antibacterial properties against MRSA and PAO-1. The HASiGdCe coating has been shown to have superior osteogenic and angiogenic properties compared to the HA coating in in vitro cellular experiments. Animal implantation experiments have shown that the HASiGdCe coating also has excellent biosafety, antimicrobial and osteogenic properties in vivo. Conclusion The HASiGdCe coating confers excellent antibacterial, angiogenic and osteogenic properties on titanium implants, which can effectively enhance implant osseointegration and prevent bacterial infections, and it accordingly has promising applications in the treatment of bone defects related to orthopedic and dental sciences.
Collapse
Affiliation(s)
- Xinlin Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, People’s Republic of China
| | - Baojuan Yang
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, 250061, People’s Republic of China
- National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan, 250061, People’s Republic of China
| | - Mengfei Xu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, People’s Republic of China
| | - Fangyi Li
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, 250061, People’s Republic of China
- National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan, 250061, People’s Republic of China
| | - Zhaoli Geng
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, People’s Republic of China
| | - Weiqiang Cui
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, 250061, People’s Republic of China
- National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan, 250061, People’s Republic of China
| | - Xingfu Sun
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, 250061, People’s Republic of China
- National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan, 250061, People’s Republic of China
| | - Yanle Li
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan, 250061, People’s Republic of China
- National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan, 250061, People’s Republic of China
| | - Yi Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, 250012, People’s Republic of China
| |
Collapse
|
28
|
Nabil Sulyiman S, El-Rashidy AA, El Moshy S, Abbas MMS, Waly G. Nano eggshell-based slurry as a direct pulp-capping material: In vitro characterization and histopathological assessment in an experimental animal model. Int Endod J 2023; 56:1129-1146. [PMID: 37358385 DOI: 10.1111/iej.13949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
AIM Pulp vitality is essential for tooth integrity. Following pulp exposure, choosing a suitable pulp-capping material is crucial to maintain pulp vitality. However, the reparative dentine bridge created by calcium hydroxide (Ca(OH)2 ) is generally porous and incomplete. The aim of the current study is to assess the in vitro and in vivo bioactivities of nano eggshell-based slurry (NES), using NES as a direct pulp-capping material, compared with Ca(OH)2 in rabbit animal model. METHODOLOGY Nano eggshell powder (NE) was characterized for particle morphology, chemical composition and ion release. In vitro bioactivity was tested by immersion in simulated body fluid (SBF) for 7 days. For histopathological evaluation, 36 adult New Zealand rabbits (72 pulp exposures) were divided into nine groups (n = 8) according to the pulp-capping material (NES, Ca(OH)2 and no capping as negative control group) and the animals were sacrificed after 7, 14 or 28 days. The pulps of the two lower central incisors were exposed and then directly capped by Ca(OH)2 or NES or left untreated. The cavities were then sealed with glass ionomer cement. Teeth were collected for histopathological evaluation using an optical microscope. Pulp haemorrhage, inflammation, fibrosis and calcific bridge formation were assessed. Results were statistically analysed using anova and Tukey's tests. RESULTS Nano eggshell particles were spherical with a 20 nm diameter and were composed mainly of calcite. Statistical analysis showed that there was a significant increase in the release of all investigated ions between days 1 and 28, except for copper. NES group showed a significantly higher release of all elements as compared to Ca(OH)2 . Environmental scanning electron microscope micrographs of NES incubated for 7 days in SBF showed the formation of HAp with a Ca/P ratio (1.686). For histopathological evaluation, the difference between groups was statistically significant. At day 28, 75% of the pulps of the Ca(OH)2 group showed mild calcific bridge in comparison with 100% moderate calcific bridge in the NES group. The NES group showed significantly less inflammation at days 7 and 28, and higher fibrosis at day 7 compared with Ca(OH)2 . CONCLUSIONS Nano eggshell-based slurry represents a promising novel direct pulp-capping material with favourable pulp tissue response.
Collapse
Affiliation(s)
| | - Aiah A El-Rashidy
- Biomaterials Department, Faculty of Dentistry, Cairo university, Cairo, Egypt
| | - Sara El Moshy
- Oral Biology Department, Faculty of Dentistry, Cairo university, Cairo, Egypt
| | - Marwa M S Abbas
- Oral Biology Department, Faculty of Dentistry, Cairo university, Cairo, Egypt
| | - Gihan Waly
- Biomaterials Department, Faculty of Dentistry, Cairo university, Cairo, Egypt
| |
Collapse
|
29
|
Lagopati N, Pippa N, Gatou MA, Papadopoulou-Fermeli N, Gorgoulis VG, Gazouli M, Pavlatou EA. Marine-Originated Materials and Their Potential Use in Biomedicine. APPLIED SCIENCES 2023; 13:9172. [DOI: 10.3390/app13169172] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2023]
Abstract
Aquatic habitats cover almost 70% of the Earth, containing several species contributing to marine biodiversity. Marine and aquatic organisms are rich in chemical compounds that can be widely used in biomedicine (dentistry, pharmacy, cosmetology, etc.) as alternative raw biomaterials or in food supplements. Their structural characteristics make them promising candidates for tissue engineering approaches in regenerative medicine. Thus, seaweeds, marine sponges, arthropods, cnidaria, mollusks, and the biomaterials provided by them, such as alginate, vitamins, laminarin, collagen, chitin, chitosan, gelatin, hydroxyapatite, biosilica, etc., are going to be discussed focusing on the biomedical applications of these marine-originated biomaterials. The ultimate goal is to highlight the sustainability of the use of these biomaterials instead of conventional ones, mainly due to the antimicrobial, anti-inflammatory, anti-aging and anticancer effect.
Collapse
Affiliation(s)
- Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Maria-Anna Gatou
- Laboratory of General Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15772 Athens, Greece
| | - Nefeli Papadopoulou-Fermeli
- Laboratory of General Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15772 Athens, Greece
| | - Vassilis G. Gorgoulis
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Clinical Molecular Pathology, Medical School, University of Dundee, Dundee DD1 9SY, UK
- Molecular and Clinical Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- School of Science and Technology, Hellenic Open University, 26335 Patra, Greece
| | - Evangelia A. Pavlatou
- Laboratory of General Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15772 Athens, Greece
| |
Collapse
|
30
|
Shirosaki Y, Fregnan F, Muratori L, Yasutomi S, Geuna S, Raimondo S. The Impact of the Molecular Weight of Degradation Products with Silicon from Porous Chitosan-Siloxane Hybrids on Neuronal Cell Behavior. Polymers (Basel) 2023; 15:3272. [PMID: 37571166 PMCID: PMC10422348 DOI: 10.3390/polym15153272] [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: 07/04/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Silicon (Si) is an essential trace element in the human body and it exists in connective tissue as aqueous orthosilicic acid. Porous chitosan-3-glycidoxypropyltrimethoxysilane (GPTMS) hybrids can regenerate nerve tissue and recover sensor and motor functions. However, the structures and roles of the degradation products with Si extracted from the hybrids in nerve regeneration are not clear. In this study, we prepared porous chitosan-GPTMS hybrids with different amounts of GPTMS to amino groups of chitosan (chitosan:GPTMS = 1:0.5 and 1:1 molar ratios). The structures of the degradation products with Si from the hybrids were examined using time-of-flight mass spectrometry, and biological assessments were conducted in order to evaluate their potential use in the preparation of devices for nerve repair. Glial and motor cell lines and ex vivo explants of dorsal root ganglia were used in this study for evaluating their behavior in the presence of the different degradation products with Si. The structure of the degradation products with Si depended on the starting composition. The results showed that glial cell proliferation was lower in the medium with the higher-molecular-weight degradation products with Si. Moreover, motor cell line differentiation and the neurite outgrowth of dorsal root ganglion explants were improved with the lower-molecular-weight degradation products with Si. The results obtained could be useful for designing a new nerve regeneration scaffold including silicon components.
Collapse
Affiliation(s)
- Yuki Shirosaki
- Faculty of Engineering, Kyushu Institute of Technology, 1-1 Sensui-cho, Tobata-ku, Kitakyushu 804-8550, Japan
| | - Federica Fregnan
- Department of Clinical and Biological Sciences and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (F.F.); (L.M.); (S.G.); (S.R.)
| | - Luisa Muratori
- Department of Clinical and Biological Sciences and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (F.F.); (L.M.); (S.G.); (S.R.)
| | - Saki Yasutomi
- Faculty of Engineering, Kyushu Institute of Technology, 1-1 Sensui-cho, Tobata-ku, Kitakyushu 804-8550, Japan
| | - Stefano Geuna
- Department of Clinical and Biological Sciences and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (F.F.); (L.M.); (S.G.); (S.R.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (F.F.); (L.M.); (S.G.); (S.R.)
| |
Collapse
|
31
|
Kato G, Araújo R, Rodrigues C, Gomes PS, Grenho L, Fernandes MH. Ex Vivo Osteogenesis Induced by Calcium Silicate-Based Cement Extracts. J Funct Biomater 2023; 14:314. [PMID: 37367277 DOI: 10.3390/jfb14060314] [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: 04/25/2023] [Revised: 05/27/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
Calcium silicate-based cements are used in a variety of clinical conditions affecting the pulp tissue, relying on their inductive effect on tissue mineralization. This work aimed to evaluate the biological response of calcium silicate-based cements with distinct properties-the fast-setting Biodentine™ and TotalFill® BC RRM™ Fast Putty, and the classical slow-setting ProRoot® MTA, in an ex vivo model of bone development. Briefly, eleven-day-old embryonic chick femurs were cultured for 10 days in organotypic conditions, being exposed to the set cements' eluates and, at the end of the culture period, evaluated for osteogenesis/bone formation by combining microtomographic analysis and histological histomorphometric assessment. ProRoot® MTA and TotalFill® extracts presented similar levels of calcium ions, although significantly lower than those released from BiodentineTM. All extracts increased the osteogenesis/tissue mineralization, assayed by microtomographic (BV/TV) and histomorphometric (% of mineralized area; % of total collagen area, and % of mature collagen area) indexes, although displaying distinct dose-dependent patterns and quantitative values. The fast-setting cements displayed better performance than that of ProRoot® MTA, with BiodentineTM presenting the best performance, within the assayed experimental model.
Collapse
Affiliation(s)
- Gabriel Kato
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| | - Rita Araújo
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| | - Cláudia Rodrigues
- Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
| | - Pedro Sousa Gomes
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| | - Liliana Grenho
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| | - Maria Helena Fernandes
- Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, 4200-393 Porto, Portugal
- LAQV/Requimte, University of Porto, 4100-007 Porto, Portugal
| |
Collapse
|
32
|
Bioglass obtained via one-pot synthesis as osseointegrative drug delivery system. Int J Pharm 2023; 633:122610. [PMID: 36669580 DOI: 10.1016/j.ijpharm.2023.122610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Osseointegration is a fundamental process during which implantable biomaterial integrates with host bone tissue. The surgical procedure of biomaterial implantation is highly associated with the risk of bacterial infection. Thus, the research continues for biodegradable bone void fillers which are able to stimulate the bone tissue regeneration and locally deliver the antibacterial agent. Herein, we obtained bifunctional bioglass (BG) using novel, preoptimized, rapid one-pot synthesis. Following the ISO Standards, the influence of the obtained BG on osteoblast-mediated phenomena, such as osteoconduction and osteoinduction was assessed and compared to two commercial materials: bioactive glass powder 45S and bioactive glass powder 85S. Direct-contact tests revealed osteoblast adhesion to BG particles; whereas, tests on extracts confirmed high viability of cells incubated with BG extract. Analyses of gene expression, alkaline phosphatase activity, and calcium phosphates deposition confirmed the stimulation of early and late stages of osteoblast differentiation and mineralization. Additionally, an extended evaluation of intracellular calcium fluctuations revealed a possible correlation between osteoblast calcium uptake and extracellular matrix mineralization. Moreover, proposed bioglass exhibited satisfactory doxycycline adsorption capacity and release profile. The obtained results confirmed the bifunctionality of the proposed BG and indicated its potential as osseointegrative bone drug delivery system.
Collapse
|
33
|
Ojstršek A, Chemelli A, Osmić A, Gorgieva S. Dopamine-Assisted Modification of Polypropylene Film to Attain Hydrophilic Mineral-Rich Surfaces. Polymers (Basel) 2023; 15:polym15040902. [PMID: 36850186 PMCID: PMC9962719 DOI: 10.3390/polym15040902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The presented study focuses on the modification of polypropylene (PP) film with tetraethyl orthosilicate (TEOS) under heterogeneous conditions via polydopamine/polyethylene imine (PDA/PEI) chemistry using a facile dip-coating procedure to attain hydrophilic mineral-rich surfaces. Thus, the resulting PP-based films were further immersed in ion-rich simulated body fluid (SBF) to deposit Ca-based minerals onto the film's surfaces efficiently. In addition, the chemical reaction mechanism on PP film was proposed, and mineralisation potential inspected by determination of functional groups of deposits, zeta potential, hydrophilicity and surface morphology/topography using Fourier transform infrared (FTIR) spectroscopy, streaming potential, water contact angle (WCA), scanning electron microscopy (SEM) and atomic force microscopy (AFM). The obtained results show the improved wettability of samples on account of PDA inclusion (WCA was reduced from 103° for pure PP film to 28° for PDA-modified film), as well as the presence of functional groups, due to the PDA/PEI/TEOS surface functionalisation, increased the ability of minerals to nucleate on the PP film's surface when it was exposed to an SBF medium. Moreover, the higher surface roughness due to the silica coatings influenced the enhanced anchoring and attachment of calcium phosphate (CaP), revealing the potential of such a facile approach to modify the chemically inert PP films, being of particular interest in different fields, including regenerative medicine.
Collapse
Affiliation(s)
- Alenka Ojstršek
- Institute of Engineering Materials and Design, Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Angela Chemelli
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - Azra Osmić
- Institute for Automation, Faculty of Electrical Engineering and Computer Science, University of Maribor, Koroška cesta 46, 2000 Maribor, Slovenia
| | - Selestina Gorgieva
- Institute of Engineering Materials and Design, Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Correspondence: ; Tel.: +386-2-220-7740
| |
Collapse
|
34
|
Cai Z, Guo Z, Yang C, Wang F, Zhang P, Wang Y, Guo M, Wang Z, Huang J, Zhang L. Surface Biofunctionalization of Gadolinium Phosphate Nanobunches for Boosting Osteogenesis/Chondrogenesis Differentiation. Int J Mol Sci 2023; 24:ijms24032032. [PMID: 36768355 PMCID: PMC9917229 DOI: 10.3390/ijms24032032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
In order to achieve smart biomedical micro/nanomaterials, promote interaction with biomolecules, improve osteogenic/chondrogenic differentiation, exhibit better dispersion in bone implants and ultimately maximize functionality, we innovatively and successfully designed and synthesized polymer PBLG-modified GdPO4·H2O nanobunches by hydroxylation, silylation and glutamylation processes. The effects of different feeding ratios on the surface coating of GdPO4·H2O with Si-OH, the grafting γ-aminopropyltriethoxysilane (APS) and the in situ ring-opening polymerization reaction of poly(g-benzyl-L-glutamate) (PBLG) were investigated, and the physical and chemical properties were characterized in detail. When GdPO4·H2O@SiO2-APS:NCA = 4:1, the PBLG-g-GdPO4·H2O grafting rate was 5.93%, with good stability and dispersion in degradable polymeric materials. However, the MRI imaging signal was sequentially weakened as the modification process proceeded. Despite this, the biological effects had surprising findings. All the modifiers at appropriate concentrations were biocompatible and biologically active and the biomacromolecules of COL I and COL II in particular were expressed at least 3 times higher in GdPO4·H2O@SiO2 compared to the PLGA. This indicates that the appropriate surface modification and functionalization of gadolinium-containing micro/nanomaterials can promote interaction with cells and encourage bone regeneration by regulating biomacromolecules and can be used in the field of biomedical materials.
Collapse
Affiliation(s)
- Zhongxing Cai
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- School of Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Ziyi Guo
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- School of Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Chaohui Yang
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- School of Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Fei Wang
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- School of Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Min Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jing Huang
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- School of Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
- Correspondence: (J.H.); (L.Z.)
| | - Long Zhang
- School of Chemical Engineering, Changchun University of Technology, Changchun 130012, China
- Correspondence: (J.H.); (L.Z.)
| |
Collapse
|
35
|
Eugen G, Claus M, Anna-Maria S, Niklas D, Philipp S, Andrea E, Andrea ML, Elke V. Degradation of 3D-printed magnesium phosphate ceramics in vitro and a prognosis on their bone regeneration potential. Bioact Mater 2023; 19:376-391. [PMID: 35574054 PMCID: PMC9062425 DOI: 10.1016/j.bioactmat.2022.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/30/2022] [Accepted: 04/17/2022] [Indexed: 12/13/2022] Open
Abstract
Regenerative bone implants promote new bone formation and ideally degrade simultaneously to osteogenesis. Although clinically established calcium phosphate bone grafts provide excellent osseointegration and osteoconductive efficacy, they are limited in terms of bioresorption. Magnesium phosphate (MP) based ceramics are a promising alternative, because they are biocompatible, mechanically extremely stable, and degrade much faster than calcium phosphates under physiological conditions. Bioresorption of an implant material can include both chemical dissolution as well as cellular resorption. We investigated the bioresorption of 3D powder printed struvite and newberyite based MP ceramics in vitro by a direct human osteoclast culture approach. The osteoclast response and cellular resorption was evaluated by means of fluorescence and TRAP staining, determination of osteoclast activities (CA II and TRAP), SEM imaging as well as by quantification of the ion release during cell culture. Furthermore, the bioactivity of the materials was investigated via SBF immersion, whereas hydroxyapatite precipitates were analyzed by SEM and EDX measurements. This bioactive coating was resorbed by osteoclasts. In contrast, only chemical dissolution contributed to bioresorption of MP, while no cellular resorption of the materials was observed. Based on our results, we expect an increased bone regeneration effect of MP compared to calcium phosphate based bone grafts and complete chemical degradation within a maximum of 1.5-3.1 years.
Collapse
Affiliation(s)
- Gefel Eugen
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| | - Moseke Claus
- Institute for Biomedical Engineering (IBMT), University of Applied Sciences Mittelhessen (THM), Wiesenstraße 14, Gießen, Germany
| | - Schmitt Anna-Maria
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| | - Dümmler Niklas
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| | - Stahlhut Philipp
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| | - Ewald Andrea
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| | - Meyer-Lindenberg Andrea
- Clinic for Small Animal Surgery and Reproduction, Ludwig-Maximilians-Universität, Munich, Germany
| | - Vorndran Elke
- Institute and Department for Functional Materials in Medicine and Dentistry, University Clinic Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
36
|
Oryan A, Hassanajili S, Sahvieh S. Zoledronate loaded polylactic acid/polycaprolactone/hydroxyapatite scaffold accelerates regeneration and led to enhance structural performance and functional ability of the radial bone defect in rat. IRANIAN JOURNAL OF VETERINARY RESEARCH 2023; 24:122-125. [PMID: 37790115 PMCID: PMC10542868 DOI: 10.22099/ijvr.2023.43807.6421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/22/2022] [Accepted: 01/24/2023] [Indexed: 10/05/2023]
Abstract
Abstract. Background One of the most common concerns in the regeneration of massive bone defects necessitating surgery and bone grafts is the application of tissue engineering using drug delivery. Zoledronate is a well-known effective drug for the healing bone fractures in osteoporotic patients. Aims An attempt was made to design a more efficient bone scaffold with polycaprolactone, polylactic acid, and hydroxyapatite. Methods The scaffold was fabricated by freeze-drying and indirect 3D printing approaches. X-ray diffraction, Fourier transform infrared spectroscopy, rheometry, scanning electron microscopy, and neutral red tests were performed to characterize the scaffold. qRT-PCR was also done to define the osteoinductivity and angiogenic induction capacity of this scaffold. Forty rats were selected and randomly divided into four groups: the control group, which received no treatment, the autograft group, scaffold group, and Zol-loaded scaffold group (n=10 in each group). The injured area was studied by radiology, biomechanical analysis, histopathology, histomorphometry, immunohistochemistry, and CT scan analyses. Results The qRT-PCR results demonstrated significantly higher expression levels of OPN, OCN, and CD31 markers in the scaffold group when compared to the control group (P<0.05). Histopathologically, the newly formed bone tissue was significantly detected in the Zol-loaded scaffold and autograft groups in comparison with the non-treated group (P<0.001). The immunohistochemistry (OC marker), biomechanical, and histomorphometric results indicated a significant improvement in the regeneration of the injured area in the groups treated with autologous bone and Zol-loaded scaffold compared to the non-treated group (P<0.05). Conclusion: The Zol-loaded scaffold accelerated bone regeneration, and led to enhanced structural performance and functional ability of the injured radial bone in rats.
Collapse
Affiliation(s)
- A. Oryan
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - S. Hassanajili
- Department of Chemical Engineering, School of Chemical and Petroleum Engineering, Shiraz University, Shiraz, Iran
| | - S. Sahvieh
- Graduated from School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
37
|
Qiu L, Zhang C, Yang X, Peng F, Huang Y, He Y. A SiO 2 layer on PEO-treated Mg for enhanced corrosion resistance and bone regeneration. Front Bioeng Biotechnol 2022; 10:1053944. [PMID: 36619395 PMCID: PMC9816664 DOI: 10.3389/fbioe.2022.1053944] [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: 10/13/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Magnesium (Mg) is a promising biodegradable metal for orthopedic applications, and plasma electrolytic oxidation (PEO) has been widely studied as a corrosion protection coating on Mg-based implants. However, the porous structures and easily formed cracks in fluid are disadvantageous for long-term corrosion protection. In this study, a SiO2 layer was deposited on PEO-treated Mg to inhibit the formation of cracks on the PEO layer and prevent the permeation of corrosive fluid. The SiO2 layer did not alter the surface morphology of the PEO layer but considerably enhanced its corrosion resistance. The in vitro culture of MC3T3-E1 cells demonstrated the good cytocompatibility and osteogenic induction ability of SiO2-coated PEO-treated Mg, which could be attributed to Mg and Si ions released from the coating. The coating also favored the angiogenesis behaviors of HUVEC. Furthermore, with the continuous release of Mg and Si ions, the as-prepared implant showed a superior osseointegration ability in a rat bone implantation model. In summary, this newly designed Mg-based implant shows promising potential for orthopedic applications.
Collapse
Affiliation(s)
- Longhai Qiu
- Department of Traumatology and Orthopaedic Surgery, Institute of Orthopaedics, Huizhou Central People’s Hospital, Huizhou, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chi Zhang
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoming Yang
- Department of Orthopaedics, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Feng Peng
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuliang Huang
- Department of Traumatology and Orthopaedic Surgery, Institute of Orthopaedics, Huizhou Central People’s Hospital, Huizhou, China,*Correspondence: Yuliang Huang, ; Yue He,
| | - Yue He
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China,*Correspondence: Yuliang Huang, ; Yue He,
| |
Collapse
|
38
|
Enhanced corrosion resistance in an inflammatory environment and osteogenic properties of silicalite-1 coated titanium alloy implants. Colloids Surf B Biointerfaces 2022; 220:112922. [DOI: 10.1016/j.colsurfb.2022.112922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
|
39
|
Inorganic/Biopolymers Hybrid Hydrogels Dual Cross-Linked for Bone Tissue Regeneration. Gels 2022; 8:gels8120762. [PMID: 36547286 PMCID: PMC9777565 DOI: 10.3390/gels8120762] [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: 11/04/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
In tissue engineering, the potential of re-growing new tissue has been considered, however, developments towards such clinical and commercial outcomes have been modest. One of the most important elements here is the selection of a biomaterial that serves as a "scaffold" for the regeneration process. Herein, we designed hydrogels composed of two biocompatible natural polymers, namely gelatin with photopolymerizable functionalities and a pectin derivative amenable to direct protein conjugation. Aiming to design biomimetic hydrogels for bone regeneration, this study proposes double-reinforcement by way of inorganic/biopolymer hybrid filling composed of Si-based compounds and cellulose nanofibers. To attain networks with high flexibility and elastic modulus, a double-crosslinking strategy was envisioned-photochemical and enzyme-mediated conjugation reactions. The dual cross-linked procedure will generate intra- and intermolecular interactions between the protein and polysaccharide and might be a resourceful strategy to develop innovative scaffolding materials.
Collapse
|
40
|
Trius-Soler M, Tresserra-Rimbau A, Moreno JJ, Peris P, Estruch R, Lamuela-Raventós RM. Effect of moderate beer consumption (with and without ethanol) on osteoporosis in early postmenopausal women: Results of a pilot parallel clinical trial. Front Nutr 2022; 9:1014140. [PMID: 36458171 PMCID: PMC9705783 DOI: 10.3389/fnut.2022.1014140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/31/2022] [Indexed: 01/08/2025] Open
Abstract
INTRODUCTION Osteoporosis is a chronic progressive bone disease characterized by low bone mineral density (BMD) and micro-architectural deterioration of bone tissue, leading to an increase in bone fragility and the risk of fractures. A well-known risk factor for bone loss is postmenopausal status. Beer may have a protective effect against osteoporosis associated with its content of silicon, polyphenols, iso-α-acids and ethanol, and its moderate consumption may therefore help to reduce bone loss in postmenopausal women. METHODS Accordingly, a 2-year controlled clinical intervention study was conducted to evaluate if a moderate daily intake of beer with (AB) or without alcohol (NAB) could have beneficial effects on bone tissue. A total of 31 postmenopausal women were assigned to three study groups: 15 were administered AB (330 mL/day) and six, NAB (660 mL/day), whereas, the 10 in the control group refrained from consuming alcohol, NAB, and hop-related products. At baseline and subsequent assessment visits, samples of plasma and urine were taken to analyze biochemical parameters, and data on medical history, diet, and exercise were collected. BMD and the trabecular bone score (TBS) were determined by dual-energy X-ray absorptiometry. Markers of bone formation (bone alkaline phosphatase [BAP] and N-propeptide of type I collagen [PINP]) and bone resorption (N-telopeptide of type I collagen [NTX] and C-telopeptide of type I collagen [CTX]) were determined annually. RESULTS Bone formation markers had increased in the AB and NAB groups compared to the control after the 2-year intervention. However, the evolution of BMD and TBS did not differ among the three groups throughout the study period. DISCUSSION Therefore, according to the findings of this pilot study, moderate beer intake does not seem to have a protective effect against bone loss in early post-menopausal women.
Collapse
Affiliation(s)
- Marta Trius-Soler
- Department of Nutrition, Food Sciences and Gastronomy, XIA, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA), University of Barcelona, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Tresserra-Rimbau
- Department of Nutrition, Food Sciences and Gastronomy, XIA, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA), University of Barcelona, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan J. Moreno
- Department of Nutrition, Food Sciences and Gastronomy, XIA, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA), University of Barcelona, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Peris
- Rheumatology Department, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Ramon Estruch
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Rosa M. Lamuela-Raventós
- Department of Nutrition, Food Sciences and Gastronomy, XIA, School of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Nutrition and Food Safety Research Institute (INSA), University of Barcelona, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Synthesis and Evaluation of a Chitosan–Silica-Based Bone Substitute for Tissue Engineering. Int J Mol Sci 2022; 23:ijms232113379. [DOI: 10.3390/ijms232113379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Bone defects have prompted the development of biomaterial-based bone substitutes for restoring the affected tissue completely. Although many biomaterials have been designed and evaluated, the combination of properties required in a biomaterial for bone tissue engineering still poses a challenge. In this study, a chitosan–silica-based biocomposite was synthetized, and its physicochemical characteristics and biocompatibility were characterized, with the aim of exploring the advantages and drawbacks of its use in bone tissue engineering. Dynamic light scattering measurements showed that the mean hydrodynamic size of solid silica particles (Sol-Si) was 482 ± 3 nm. Scanning electron microscopy of the biocomposite showed that Sol-Si were homogenously distributed within the chitosan (CS) matrix. The biocomposite swelled rapidly and was observed to have no cytotoxic effect on the [3T3] cell line within 24 h. Biocompatibility was also analyzed in vivo 14 days post-implant using a murine experimental model (Wistar rats). The biocomposite was implanted in the medullary compartment of both tibiae (n = 12). Histologically, no acute inflammatory infiltrate or multinucleated giant cells associated to the biocomposite were observed, indicating good biocompatibility. At the tissue–biocomposite interface, there was new formation of woven bone tissue in close contact with the biocomposite surface (osseointegration). The new bone formation may be attributed to the action of silica. Free silica particles originating from the biocomposite were observed at the tissue–biocomposite interface. According to our results, the biocomposite may act as a template for cellular interactions and extracellular matrix formation, providing a structural support for new bone tissue formation. The CS/Sol-Si biocomposite may act as a Si reservoir, promoting new bone formation. A scaffold with these properties is essential for cell differentiation and filling a bone defect.
Collapse
|
42
|
The Influence of the Matrix on the Apatite-Forming Ability of Calcium Containing Polydimethylsiloxane-Based Cements for Endodontics. Molecules 2022; 27:molecules27185750. [PMID: 36144487 PMCID: PMC9504520 DOI: 10.3390/molecules27185750] [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: 07/31/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
This study aimed to characterize the chemical properties and bioactivity of an endodontic sealer (GuttaFlow Bioseal) based on polydimethylsiloxane (PDMS) and containing a calcium bioglass as a doping agent. Commercial PDMS-based cement free from calcium bioglass (GuttaFlow 2 and RoekoSeal) were characterized for comparison as well as GuttaFlow 2 doped with dicalcium phosphate dihydrate, hydroxyapatite, or a tricalcium silicate-based cement. IR and Raman analyses were performed on fresh materials as well as after aging tests in Hank’s Balanced Salt Solution (28 d, 37 °C). Under these conditions, the strengthening of the 970 cm−1 Raman band and the appearance of the IR components at 1455−1414, 1015, 868, and 600−559 cm−1 revealed the deposition of B-type carbonated apatite. The Raman I970/I638 and IR A1010/A1258 ratios (markers of apatite-forming ability) showed that bioactivity decreased along with the series: GuttaFlow Bioseal > GuttaFlow 2 > RoekoSeal. The PDMS matrix played a relevant role in bioactivity; in GuttaFlow 2, the crosslinking degree was favorable for Ca2+ adsorption/complexation and the formation of a thin calcium phosphate layer. In the less crosslinked RoekoSeal, such processes did not occur. The doped cements showed bioactivity higher than GuttaFlow 2, suggesting that the particles of the mineralizing agents are spontaneously exposed on the cement surface, although the hydrophobicity of the PDMS matrix slowed down apatite deposition. Relevant properties in the endodontic practice (i.e., setting time, radiopacity, apatite-forming ability) were related to material composition and the crosslinking degree.
Collapse
|
43
|
Mohammed EEA, Beherei HH, El-Zawahry M, Farrag ARH, Kholoussi N, Helwa I, Mabrouk M, Abdel Aleem AK. Osteogenic enhancement of modular ceramic nanocomposites impregnated with human dental pulp stem cells: an approach for bone repair and regenerative medicine. J Genet Eng Biotechnol 2022; 20:123. [PMID: 35976537 PMCID: PMC9385929 DOI: 10.1186/s43141-022-00387-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Background/aim Human dental pulp-derived mesenchymal stem cells (hDP-MSCs) are a promising source of progenitor cells for bone tissue engineering. Nanocomposites made of calcium phosphate especially hydroxyapatite (HA) offer an impressive solution for orthopedic and dental implants. The combination of hDP-MSCs and ceramic nanocomposites has a promising therapeutic potential in regenerative medicine. Despite the calcium phosphate hydroxyapatite (HA)-based nanocomposites offer a good solution for orthopedic and dental implants, the heavy load-bearing clinical applications require higher mechanical strength, which is not of the HA’ properties that have low mechanical strength. Herein, the outcomes of using fabricated ceramic nanocomposites of hydroxyapatite/titania/calcium silicate mixed at different ratios (C1, C2, and C3) and impregnated with hDP-MSCs both in in vitro cultures and rabbit model of induced tibial bone defect were investigated. Our aim is to find out a new approach that would largely enhance the osteogenic differentiation of hDP-MSCs and has a therapeutic potential in bone regeneration. Subjects and methods Human DP-MSCs were isolated from the dental pulp of the third molar and cultured in vitro. Alizarin Red staining was performed at different time points to assess the osteogenic differentiation. Flow cytometer was used to quantify the expression of hDP-MSCs unique surface markers. Rabbits were used as animal models to evaluate the therapeutic potential of osteogenically differentiated hDP-MSCs impregnated with ceramic nanocomposites of hydroxyapatite/tatiana/calcium silicate (C1, C2, and C3). Histopathological examination and scanning electron microscopy (SEM) were performed to evaluate bone healing potential in the rabbit induced tibial defects three weeks post-transplantation. Results The hDP-MSCs showed high proliferative and osteogenic potential in vitro culture. Their osteogenic differentiation was accelerated by the ceramic nanocomposites’ scaffold and revealed bone defect’s healing in transplanted rabbit groups compared to control groups. Histopathological and SEM analysis of the transplanted hDP-MSCs/ceramic nanocomposites showed the formation of new bone filling in the defect area 3 weeks post-implantation. Accelerate osseointegration and enhancement of the bone-bonding ability of the prepared nanocomposites were also confirmed by SEM. Conclusions The results strongly suggested that ceramic nanocomposites of hydroxyapatite/ titania /calcium silicate (C1, C2, and C3) associated with hDP-MSCs have a therapeutic potential in bone healing in a rabbit model. Hence, the combined osteogenic system presented here is recommended for application in bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Eman E A Mohammed
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt. .,Refractoriness, Ceramics and Building Materials Department, Inorganic Chemical Industries and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt.
| | - Hanan H Beherei
- Fixed and Removable Prosthodontics Department, Oral and Dental Research Institute, National Research Centre, Cairo, Egypt
| | - Mohamed El-Zawahry
- Pathology Department, Medicine and Clinical Studies Research Institute, National Research Centre, Cairo, Egypt
| | - Abdel Razik H Farrag
- Stem Cell Research Group, Medical Research Center of Excellence, National Research Centre, Cairo, Egypt
| | - Naglaa Kholoussi
- Immunogenetics Department, Human Genetics and Genome Research Institute, National Research Centre, National Research Centre, Cairo, Egypt
| | - Iman Helwa
- Immunogenetics Department, Human Genetics and Genome Research Institute, National Research Centre, National Research Centre, Cairo, Egypt
| | - Mostafa Mabrouk
- Fixed and Removable Prosthodontics Department, Oral and Dental Research Institute, National Research Centre, Cairo, Egypt
| | - Alice K Abdel Aleem
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.,Refractoriness, Ceramics and Building Materials Department, Inorganic Chemical Industries and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
44
|
In Vivo Application of Silica-Derived Inks for Bone Tissue Engineering: A 10-Year Systematic Review. Bioengineering (Basel) 2022; 9:bioengineering9080388. [PMID: 36004914 PMCID: PMC9404869 DOI: 10.3390/bioengineering9080388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
As the need for efficient, sustainable, customizable, handy and affordable substitute materials for bone repair is critical, this systematic review aimed to assess the use and outcomes of silica-derived inks to promote in vivo bone regeneration. An algorithmic selection of articles was performed following the PRISMA guidelines and PICO method. After the initial selection, 51 articles were included. Silicon in ink formulations was mostly found to be in either the native material, but associated with a secondary role, or to be a crucial additive element used to dope an existing material. The inks and materials presented here were essentially extrusion-based 3D-printed (80%), and, overall, the most investigated animal model was the rabbit (65%) with a femoral defect (51%). Quality (ARRIVE 2.0) and risk of bias (SYRCLE) assessments outlined that although a large majority of ARRIVE items were “reported”, most risks of bias were left “unclear” due to a lack of precise information. Almost all studies, despite a broad range of strategies and formulations, reported their silica-derived material to improve bone regeneration. The rising number of publications over the past few years highlights Si as a leverage element for bone tissue engineering to closely consider in the future.
Collapse
|
45
|
Barium Oxide Doped Magnesium Silicate Nanopowders for Bone Fracture Healing: Preparation, Characterization, Antibacterial and In Vivo Animal Studies. Pharmaceutics 2022; 14:pharmaceutics14081582. [PMID: 36015208 PMCID: PMC9415424 DOI: 10.3390/pharmaceutics14081582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 02/01/2023] Open
Abstract
Magnesium silicate (MgS) nanopowders doped with barium oxide (BaO) were prepared by sol-gel technique, which were then implanted into a fracture of a tibia bone in rats for studying enhanced in vivo bone regeneration. The produced nanopowders were characterized using X-ray diffraction (XRD), Fourier transform infrared spectra (FTIR), scanning electron microscope with energy-dispersive X-ray spectrometry (SEM-EDX) and transmission electron microscope (TEM). Mechanical and bactericidal properties of the nanopowders were also determined. Increased crystallinity, particle diameter and surface area were found to decrease after the BaO doping without any notable alterations on their chemical integrities. Moreover, elevated mechanical and antibacterial characteristics were recognized for higher BaO doping concentrations. Our animal studies demonstrated that impressive new bone tissues were formed in the fractures while the prepared samples degraded, indicating that the osteogenesis and degradability of the BaO containing MgS samples were better than the control MgS. The results of the animal study indicated that the simultaneous bone formation on magnesium biomaterial silicate and barium MgS with completed bone healing after five weeks of implantations. The findings also demonstrated that the prepared samples with good biocompatibility and degradability could enhance vascularization and osteogenesis, and they have therapeutic potential to heal bone fractures.
Collapse
|
46
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
47
|
Saghiri MA, Vakhnovetsky J, Vakhnovetsky A. Functional role of inorganic trace elements in dentin apatite-Part II: Copper, manganese, silicon, and lithium. J Trace Elem Med Biol 2022; 72:126995. [PMID: 35605438 DOI: 10.1016/j.jtemb.2022.126995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/12/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022]
Abstract
Trace elements are recognized as being essential in dentin and bone apatite. The effects of zinc, strontium, magnesium, and iron were discussed in part I. In part II, we evaluated the functional role of copper, manganese, silicon, and lithium on dentin apatite, with critical effects on morphology, crystallinity, and solubility. An electronic search was performed on the role of these trace elements in dentin apatite from January 2000 to January 2022. The recent aspects of the relationship between four different trace elements and their critical role in the structure and mechanics of dentin were assessed. These findings show that elements play a vital role in the human body, especially in the crystalline structure of dentin apatite. Copper presents immense benefits in dental restorative biomaterials because of its importance in enhancing odontogenesis. The biological role of manganese in dentin apatite is still largely unknown, but it has gained attention for many of its broad physiological functions such as modulating osteoblast proliferation, differentiation, and metabolism in bones. The functional role of silicon in dentin apatite is similarly lacking, but findings reveal its importance in mineralization and collagen formation, making it useful for the field of restorative dentistry. Likewise, lithium was found to have important roles in dentin mineralization as well as in the formation of dentin bridges and tissues. Therefore, there is growing importance in studying the aforementioned elements in the context of dentin apatite.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Director, Biomaterial and Prosthodontics Laboratory and Assistant Professor, Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, United States; Adjunct Assistant Professor, Department of Endodontics, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, United States.
| | - Julia Vakhnovetsky
- Visiting Researcher, Sector of Angiogenesis Regenerative Medicine, Dr. Hajar Afsar Lajevardi Research Cluster (DHAL), Hackensack, NJ, United States; Pre-Dental Student, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Anna Vakhnovetsky
- Pre-Medical Student, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
48
|
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
|
49
|
Vrchovecká K, Pávková-Goldbergová M, Engqvist H, Pujari-Palmer M. Cytocompatibility and Bioactive Ion Release Profiles of Phosphoserine Bone Adhesive: Bridge from In Vitro to In Vivo. Biomedicines 2022; 10:biomedicines10040736. [PMID: 35453486 PMCID: PMC9044752 DOI: 10.3390/biomedicines10040736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/09/2022] [Accepted: 03/18/2022] [Indexed: 02/06/2023] Open
Abstract
One major challenge when developing new biomaterials is translating in vitro testing to in vivo models. We have recently shown that a single formulation of a bone tissue adhesive, phosphoserine modified cement (PMC), is safe and resorbable in vivo. Herein, we screened many new adhesive formulations, for cytocompatibility and bioactive ion release, with three cell lines: MDPC23 odontoblasts, MC3T3 preosteoblasts, and L929 fibroblasts. Most formulations were cytocompatible by indirect contact testing (ISO 10993-12). Formulations with larger amounts of phosphoserine (>50%) had delayed setting times, greater ion release, and cytotoxicity in vitro. The trends in ion release from the adhesive that were cured for 24 h (standard for in vitro) were similar to release from the adhesives cured only for 5−10 min (standard for in vivo), suggesting that we may be able to predict the material behavior in vivo, using in vitro methods. Adhesives containing calcium phosphate and silicate were both cytocompatible for seven days in direct contact with cell monolayers, and ion release increased the alkaline phosphatase (ALP) activity in odontoblasts, but not pre-osteoblasts. This is the first study evaluating how PMC formulation affects osteogenic cell differentiation (ALP), cytocompatibility, and ion release, using in situ curing conditions similar to conditions in vivo.
Collapse
Affiliation(s)
- Kateřina Vrchovecká
- Department of Pathology Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (K.V.); (M.P.-G.)
| | - Monika Pávková-Goldbergová
- Department of Pathology Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (K.V.); (M.P.-G.)
| | - Håkan Engqvist
- Department of Materials Science and Engineering, Applied Material Science, Uppsala University, 75103 Uppsala, Sweden
- Correspondence: (H.E.); (M.P.-P.)
| | - Michael Pujari-Palmer
- Department of Materials Science and Engineering, Applied Material Science, Uppsala University, 75103 Uppsala, Sweden
- Correspondence: (H.E.); (M.P.-P.)
| |
Collapse
|
50
|
Osteogenic Induction with Silicon Hydroxyapatite Using Modified Autologous Adipose Tissue-Derived Stromal Vascular Fraction: In Vitro and Qualitative Histomorphometric Analysis. MATERIALS 2022; 15:ma15051826. [PMID: 35269057 PMCID: PMC8911855 DOI: 10.3390/ma15051826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/16/2022]
Abstract
Large bone defects requiring invasive surgical procedures have long been a problem for orthopedic surgeons. Despite the use of autologous bone grafting, satisfactory results are often not achieved due to associated limitations. Biomaterials are viable alternatives and have lately been used in association with Stromal Vascular Fraction (SVF), stem cells, and signaling factors for bone tissue engineering (BTE). The objective of the current study was to assess the biocompatibility of Silicon Hydroxyapatite (Si-HA) and to improve osteogenic potential by using autologous adipose-derived SVF with Si-HA in a rabbit bone defect model. Si-HA granules synthesized using a wet precipitation method were used. They were characterized using scanning electron microscopy (SEM), Fourier transform infrared (FTIR), and X-ray diffraction (XRD). A hemolysis assay was used to assess the hemolytic effects of Si-HA, while cell viability was assessed through Alamar Blue assay using MC3T3 mouse osteoblasts. The osteogenic potential of Si-HA both alone and with enzymatically/non-enzymatically-derived SVF (modified) was performed by implantation in a rabbit tibia model followed by histomorphometric analysis and SEM of dissected bone after six weeks. The results showed that Si-HA granules were microporous and phase pure and that the addition of Silicon did not influence Si-HA phase composition. Si-HA granules were found to be non-hemolytic on the hemolysis assay and non-toxic to MC3T3 mouse osteoblasts on the Alamar Blue assay. Six weeks following implantation Si-HA showed high biocompatibility, with increased bone formation in all groups compared to control. Histologically more mature bone was formed in the Si-HA implanted along with non-enzymatically-derived modified SVF. Bone formation was observed on and around Si-HA, reflecting osseointegration. In conclusion, Si-HA is osteoconductive and promotes osteogenesis, and its use with SVF enhances osteogenesis.
Collapse
|